1
|
Majid H, Kohli S, Islam SU, Nidhi. The role of branched chain aminotransferase in the interrelated pathways of type 2 diabetes mellitus and Alzheimer's disease. J Diabetes Metab Disord 2025; 24:90. [PMID: 40151764 PMCID: PMC11936868 DOI: 10.1007/s40200-025-01597-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/23/2025] [Indexed: 03/29/2025]
Abstract
Objectives This review assessed the role of Branched-Chain Amino Acid Transaminase (BCAT) enzymes in human metabolism, and their involvement in the catabolism of branched-chain amino acids (BCAAs) and exploring the association between Type 2 Diabetes Mellitus (T2DM) and Alzheimer's disease (AD) through insulin resistance. Methods The analysis involves a comprehensive literature review of recent research findings related to BCAT enzymes, BCAA metabolism, T2DM, and AD. Relevant studies and articles were identified through systematic searches in databases such as PubMed, ScienceDirect, and other scholarly resources. Inclusion criteria encompassed research articles, reviews, and studies published in peer-reviewed journals, with a focus on human metabolism, BCAT enzymes, and the interplay between BCAA metabolism, T2DM, and AD. Results The association between T2DM and AD suggests a potential metabolic link, particularly through dysregulated BCAA metabolism leading to insulin resistance. The impact of impaired insulin signaling is implicated in brain function and the accumulation of amyloid plaques facilitated by BCAT. Conclusion The identified link between BCAT, BCAA metabolism, T2DM, and AD suggests that disruptions in BCAT levels could serve as valuable indicators for early detection of insulin resistance and cognitive impairment as observed in Type 3 Diabetes which may present a promising therapeutic target.
Collapse
Affiliation(s)
- Haya Majid
- Department of Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062 India
| | - Sunil Kohli
- Department of Medicine and Diabetes Unit, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard, New Delhi, 110062 India
| | - Sajad Ul Islam
- Department of Medicine and Diabetes Unit, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard, New Delhi, 110062 India
| | - Nidhi
- Department of Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062 India
| |
Collapse
|
2
|
Banki K, Perl A. Cell type-specific regulation of the pentose phosphate pathway during development and metabolic stress-driven autoimmune diseases: Relevance for inflammatory liver, renal, endocrine, cardiovascular and neurobehavioral comorbidities, carcinogenesis, and aging. Autoimmun Rev 2025; 24:103781. [PMID: 40010622 DOI: 10.1016/j.autrev.2025.103781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
The pathogenesis of autoimmunity is incompletely understood which limits the development of effective therapies. New compelling evidence indicates that the pentose phosphate pathway (PPP) profoundly regulate lineage development in the immune system that are influenced by genetic and environmental factors during metabolic stress underlying the development of autoimmunity. The PPP provides two unique metabolites, ribose 5-phosphate for nucleotide biosynthesis in support of cell proliferation and NADPH for protection against oxidative stress. The PPP operates two separate branches, oxidative (OxPPP) and non-oxidative (NOxPPP). While the OxPPP functions in all organisms, the NOxPPP reflects adaptation to niche-specific metabolic requirements. The OxPPP primarily depends on glucose 6-phosphate dehydrogenase (G6PD), whereas transaldolase (TAL) controls the rate and directionality of metabolic flux though the NOxPPP. G6PD is essential for normal development but its partial deficiency protects from malaria. Although men and mice lacking TAL develop normally, they exhibit liver cirrhosis progressing to hepatocellular carcinoma. Mechanistic target of rapamycin-dependent loss of paraoxonase 1 drives autoimmunity and cirrhosis in TAL deficiency, while hepatocarcinogenesis hinges on polyol pathway activation via aldose reductase (AR). Accumulated polyols, such as erythritol, xylitol, and sorbitol, which are commonly used as non-caloric sweeteners, may act as pro-inflammatory oncometabolites under metabolic stress, such as TAL deficiency. The TAL/AR axis is identified as a checkpoint of pathogenesis and target for treatment of metabolic stress-driven systemic autoimmunity with relevance for inflammatory liver, renal and cardiovascular disorders, diabetes, carcinogenesis, and aging.
Collapse
Affiliation(s)
- Katalin Banki
- Departments of Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, and Pathology, State University of New York Upstate Medical University, Norton College of Medicine, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Andras Perl
- Departments of Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, and Pathology, State University of New York Upstate Medical University, Norton College of Medicine, 750 East Adams Street, Syracuse, NY 13210, USA.
| |
Collapse
|
3
|
Wadan AHS, Moshref AS, Emam AM, Bakry YG, Khalil BO, Chaurasia A, Ibrahim RAH, Badawy T, Mehanny SS. Mitochondrial dysfunction as a key player in aggravating periodontitis among diabetic patients: review of the current scope of knowledge. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04025-x. [PMID: 40272516 DOI: 10.1007/s00210-025-04025-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 03/05/2025] [Indexed: 04/25/2025]
Abstract
Periodontitis is a prevalent inflammatory disease that leads to significant periodontal tissue destruction and compromised dental health, with its severity exacerbated in individuals with Diabetes Mellitus (DM). This review explores the complex relationship between mitochondrial dysfunction and periodontitis in diabetic patients. Recent studies indicate that the excessive production of reactive oxygen species (ROS), primarily generated by dysfunctional mitochondrial electron transport chain (ETC) complexes, contributes to oxidative stress (OS) and subsequent periodontal tissue damage. The interplay between impaired mitochondrial biogenesis, apoptosis of periodontal cells, and ROS accumulation highlights a critical area of concern in understanding the pathophysiology of diabetic periodontitis. Furthermore, altered glycemic control due to inflammatory processes associated with periodontitis may perpetuate a cyclical detriment to oral and systemic health. This review aims to highlight the mechanistic roles of mitochondrial dysfunction in the aggravation of periodontitis among diabetic patients, emphasizing further research to identify potential therapeutic targets and improve treatment efficacy for this dual pathology.
Collapse
Affiliation(s)
- Al-Hassan Soliman Wadan
- Department of Oral Biology, Faculty of Dentistry, Galala University, Galala City, Suez, Egypt.
| | | | | | | | | | - Akhilanand Chaurasia
- Department of Oral Medicine and Radiology, King George'S Medical University, Lucknow, India
| | - Reham A H Ibrahim
- Department of Oral Biology, Faculty of Dentistry, Galala University, Galala City, Suez, Egypt
| | - Tamer Badawy
- Department of Oral Biology, Faculty of Dentistry, Galala University, Galala City, Suez, Egypt
- Department of Oral Biology, Faculty of Dentistry, Cairo University, Cairo, Egypt
| | - Samah S Mehanny
- Department of Oral Biology, Faculty of Dentistry, Galala University, Galala City, Suez, Egypt
- Department of Oral Biology, Faculty of Dentistry, Cairo University, Cairo, Egypt
| |
Collapse
|
4
|
Shu H, Liao Q, Chen Z, Liang M, Zhang S, Liu J, Wu Y, Hu P, Luo M, Zhu W, Zhu X, Yang L, Yan T. Flavonoids serve as a promising therapeutic agent for ischemic stroke. Brain Res 2025; 1853:149528. [PMID: 39999903 DOI: 10.1016/j.brainres.2025.149528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 02/27/2025]
Abstract
Ischemic stroke (IS) continues to be a major public health concern and is characterized by significantly high mortality and disabling rates. Inhibiting nerve cells death and enhancing the repair of ischemic tissue are important treatment concepts for IS. Currently, the mainstream treatment strategies mainly focus on short-term care, which underscores the urgent need for novel therapeutic strategies for long-term care. Emerging data reveal that flavonoids have surfaced as promising candidates for IS patients' long-term care. Flavonoids can alleviate neuroinflammation and anti-apoptosis due to their characteristic pharmacological mechanisms. Clinical evidence suggests that long-term flavonoids intake improves IS patients' long-term outcomes. Though the effect of flavonoids in IS treatment has been explored for decades, the neuroprotective pharmacodynamics have not been well established. Thereby, the aim of current review is to summarize the pathways involved in neuroprotective effect of flavonoids. This review will also advance the potential of flavonoids as a viable clinical candidate for the treatment of IS.
Collapse
Affiliation(s)
- Hongxin Shu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qiuye Liao
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Zhihao Chen
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Mingyu Liang
- School of life sciences, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Si Zhang
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Junzhe Liu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yanze Wu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ping Hu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ming Luo
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Wenping Zhu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xingen Zhu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Li Yang
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Tengfeng Yan
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
5
|
Tashkandi AJ, Gorman A, McGoldrick Mathers E, Carney G, Yacoub A, Setyaningsih WAW, Kuburas R, Margariti A. Metabolic and Mitochondrial Dysregulations in Diabetic Cardiac Complications. Int J Mol Sci 2025; 26:3016. [PMID: 40243689 PMCID: PMC11988959 DOI: 10.3390/ijms26073016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
The growing prevalence of diabetes highlights the urgent need to study diabetic cardiovascular complications, specifically diabetic cardiomyopathy, which is a diabetes-induced myocardial dysfunction independent of hypertension or coronary artery disease. This review examines the role of mitochondrial dysfunction in promoting diabetic cardiac dysfunction and highlights metabolic mechanisms such as hyperglycaemia-induced oxidative stress. Chronic hyperglycaemia and insulin resistance can activate harmful pathways, including advanced glycation end-products (AGEs), protein kinase C (PKC) and hexosamine signalling, uncontrolled reactive oxygen species (ROS) production and mishandling of Ca2+ transient. These processes lead to cardiomyocyte apoptosis, fibrosis and contractile dysfunction. Moreover, endoplasmic reticulum (ER) stress and dysregulated RNA-binding proteins (RBPs) and extracellular vesicles (EVs) contribute to tissue damage, which drives cardiac function towards heart failure (HF). Advanced patient-derived induced pluripotent stem cell (iPSC) cardiac organoids (iPS-COs) are transformative tools for modelling diabetic cardiomyopathy and capturing human disease's genetic, epigenetic and metabolic hallmarks. iPS-COs may facilitate the precise examination of molecular pathways and therapeutic interventions. Future research directions encourage the integration of advanced models with mechanistic techniques to promote novel therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Refik Kuburas
- Wellcome Wolfson Institute of Experimental Medicine, Queens University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (A.J.T.); (A.G.); (E.M.M.); (G.C.); (A.Y.); (W.A.W.S.)
| | - Andriana Margariti
- Wellcome Wolfson Institute of Experimental Medicine, Queens University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (A.J.T.); (A.G.); (E.M.M.); (G.C.); (A.Y.); (W.A.W.S.)
| |
Collapse
|
6
|
Rehman G, Sher A, Rauf A, Shah ZA, Alsanie WF, Alamri AS, Alhomrani M, Ribaudo G. In silico and in vitro evidence on the anti-diabetic effect of grandidentatin from Salix acmophylla Boiss. Fitoterapia 2025; 183:106497. [PMID: 40139406 DOI: 10.1016/j.fitote.2025.106497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/24/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Current pharmacological treatments against diabetes do not reach optimal results in many conditions, and the attention of medicinal chemists is currently dedicated to multi-target compounds with antioxidant, anti-inflammatory and anti-diabetic properties. In this connection, medicinal plants represent valuable sources. Grandidentatin is a phenolic glycoside isolated from the leaves of Salix acmophylla Boiss., and in the current study its anti-inflammatory activity was investigated in vitro through red blood cells membrane stabilization and protein denaturation assays, and it was comparable to that of the positive control diclofenac sodium. Additionally grandidentatin was effective in the glucose uptake assay, used to assess the blood sugar lowering action, in the same concentration range of metformin. Computational studies were also enrolled to investigate the physico-chemical properties and the mechanism of action, showing that the molecule possesses the features to be defined as a "drug-like" compound, and that it may target aldose reductase as observed by docking studies.
Collapse
Affiliation(s)
- Gauhar Rehman
- Department of Zoology, Abdul Wali Khan University Mardan, Mardan, KPK, Pakistan
| | - Adil Sher
- Department of Zoology, Abdul Wali Khan University Mardan, Mardan, KPK, Pakistan
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar 23561, Khyber Pakhtunkhwa, Pakistan.
| | - Zafar Ali Shah
- School of Pharmaceutical Science, Guangzhou Medical University, China
| | - Walaa F Alsanie
- Department of Clinical Laboratory Sciences, The Faculty of Applied Medical Sciences, Taif University, Taif, Saudi Arabia; Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research, Taif University, Taif 26432, Saudi Arabia
| | - Abdulhakeem S Alamri
- Department of Clinical Laboratory Sciences, The Faculty of Applied Medical Sciences, Taif University, Taif, Saudi Arabia; Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research, Taif University, Taif 26432, Saudi Arabia
| | - Majid Alhomrani
- Department of Clinical Laboratory Sciences, The Faculty of Applied Medical Sciences, Taif University, Taif, Saudi Arabia; Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research, Taif University, Taif 26432, Saudi Arabia
| | - Giovanni Ribaudo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
7
|
Rroji M, Spahia N, Figurek A, Spasovski G. Targeting Diabetic Atherosclerosis: The Role of GLP-1 Receptor Agonists, SGLT2 Inhibitors, and Nonsteroidal Mineralocorticoid Receptor Antagonists in Vascular Protection and Disease Modulation. Biomedicines 2025; 13:728. [PMID: 40149704 PMCID: PMC11940462 DOI: 10.3390/biomedicines13030728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/03/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Atherosclerosis is a closely related complication of diabetes mellitus (DM), driven by endothelial dysfunction, inflammation, and oxidative stress. The progression of atherosclerosis is accelerated by hyperglycemia, insulin resistance, and hyperlipidemia. Novel antidiabetic agents, SGLT2 inhibitors, and GLP-1 agonists improve glycemic control and offer cardiovascular protection, reducing the risk of major adverse cardiovascular events (MACEs) and heart failure hospitalization. These agents, along with nonsteroidal mineralocorticoid receptor antagonists (nsMRAs), promise to mitigate metabolic disorders and their impact on endothelial function, oxidative stress, and inflammation. This review explores the potential molecular mechanisms through which these drugs may prevent the development of atherosclerosis and cardiovascular disease (CVD), supported by a summary of preclinical and clinical evidence.
Collapse
Affiliation(s)
- Merita Rroji
- Department of Nephrology, University of Medicine Tirana, 1001 Tirana, Albania
- Department of Nephrology, University Hospital Center Mother Tereza, 1001 Tirana, Albania;
| | - Nereida Spahia
- Department of Nephrology, University Hospital Center Mother Tereza, 1001 Tirana, Albania;
| | - Andreja Figurek
- Institute of Anatomy, University of Zurich, 8057 Zurich, Switzerland;
| | - Goce Spasovski
- Department of Nephrology, University Sts. Cyril and Methodius, 1000 Skopje, North Macedonia;
| |
Collapse
|
8
|
Mhaske PS, Nathan B, Nallusamy S, Raman R, Sampathkumar V, Kathirvel P, Ravikumar CN, Sathyaseelan C, Selvakumar D. Exploring the anti-diabetic potential of barnyard millet: insights from virtual screening, MD simulation and MM-PBSA. J Biomol Struct Dyn 2025:1-15. [PMID: 40084843 DOI: 10.1080/07391102.2025.2478466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 01/18/2025] [Indexed: 03/16/2025]
Abstract
Barnyard millet (Echinochloa frumentacea) is a nutritionally superior grain and a rich source of dietary fiber, and protein. It helps in managing health and dietary issues such as malnutrition, diabetes, obesity, and celiac disease. Its low content of slowly digestible carbohydrates promotes a gradual release of glucose, helping to maintain stable blood glucose levels. The present study aims to identify and screen phytochemicals in the barnyard millet and explore its anti-diabetic activity through an in-silico study. Gas chromatography-mass spectrometry (GC-MS) analyses of the seed extract revealed the occurrence of 73 bioactive compounds that are known to possess a variety of pharmacological activities. Based on the virtual screening analysis, phytochemicals interacted with five different diabetic targets, with diosgenin demonstrating the lowest binding affinity across four receptors. Specifically, diosgenin showed a binding affinity of -9.2 kcal/mol with the Insulin receptor (PDB ID: 1IR3), -8.7 kcal/mol with Peroxisome proliferator-activated receptors (PDB ID: 3G9E), -7.5 kcal/mol with Tyrosine phosphatase 1-beta (2F70), and -6.5 kcal/mol with the Glucagon receptor (PDB ID: 5EE7). For Aldose reductase (PDB ID: 4XZH), Docosahexaenoic acid exhibited the lowest binding affinity of -9.9 kcal/mol. The dynamic behavior of 2F70-Diosgenin docked complexes throughout a 500 ns trajectory run was investigated further. The RMSD and RMSF analyses reveal that the complex remains structurally stable. The binding free energies were computed using the Molecular Mechanics/Poisson-Boltzmann Surface Area (MM/PBSA) methodology. The calculation results show that the predicted free energies of the complex are stable. These results suggest that the 2F70-Diosgenin complex is stable, highlighting its potential for further wet lab validation.
Collapse
Affiliation(s)
- Pallavi Sukdev Mhaske
- Department of Plant Molecular Biology and Bioinformatics, CPMB&B, TNAU, Coimbatore, India
| | - Bharathi Nathan
- Department of Plant Molecular Biology and Bioinformatics, CPMB&B, TNAU, Coimbatore, India
| | - Saranya Nallusamy
- Department of Plant Molecular Biology and Bioinformatics, CPMB&B, TNAU, Coimbatore, India
| | - Renuka Raman
- Department of Plant Biotechnology, CPMB&B, TNAU, Coimbatore, India
| | | | - Pavitra Kathirvel
- Department of Plant Molecular Biology and Bioinformatics, CPMB&B, TNAU, Coimbatore, India
| | | | - Chakkarai Sathyaseelan
- Department of Plant Molecular Biology and Bioinformatics, CPMB&B, TNAU, Coimbatore, India
| | - Divya Selvakumar
- Department of Plant Molecular Biology and Bioinformatics, CPMB&B, TNAU, Coimbatore, India
| |
Collapse
|
9
|
Reddy KTK, Rakesh K, Prathyusha S, Gupta JK, Nagasree K, Lokeshvar R, Elumalai S, Prasad PD, Kolli D. Revolutionizing Diabetes Care: The Role of Marine Bioactive Compounds and Microorganisms. Cell Biochem Biophys 2025; 83:193-213. [PMID: 39254792 DOI: 10.1007/s12013-024-01508-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2024] [Indexed: 09/11/2024]
Abstract
Diabetes is a metabolic condition characterized by high blood glucose levels. Aquatic products like microalgae, bacteria, seagrasses, macroalgae, corals, and sponges have been investigated for potential anti-diabetic properties. We looked at polyphenols, peptides, pigments, and sterols, as well as other bioactive substances found in marine resources, to see if they could help treat or manage diabetes, in addition to describing the several treatment strategies that alter diabetes and its implications, such as inhibition of protein tyrosine phosphatases 1B (PTP1B), α-glucosidase, α-amylase, dipeptidyl peptidase IV (DPP-IV), aldose reductase, lipase, glycogen synthase kinase 3β (GSK-3β), and insulin resistance prevention, promotion of liver antioxidant capacity, natural killer cell stimulant, anti-inflammatory actions, increased AMP-activated protein kinase (AMPK) phosphorylation and sugar and metabolism of the lipid, reducing oxidative stress, and β-pancreatic cell prevention. This study highlights the revolutionary potential of marine bioactive compounds and microorganisms in transforming diabetes care. We believe in a future in which innovative, sustainable, and efficient therapeutic approaches will result in improved quality of life and better outcomes for people with diabetes mellitus by forging a new path for treatment, utilizing the power of the world's oceans, and capitalizing on the symbiotic relationship between humans and the marine ecosystem. This study area offers optimism and promising opportunities for transforming diabetes care.
Collapse
Affiliation(s)
- Konatham Teja Kumar Reddy
- Department of Pharmacy, University College of Technology, Osmania University, Amberpet, Hyderabad, Telangana, India
| | - Kamsali Rakesh
- Department of Chemistry, Koneru Lakshmaiah Education Foundation, Greenfields, Vaddeswaram, Guntur, Andhra Pradesh, India
| | - Segu Prathyusha
- Department of Pharmacognosy, School of Pharmacy, Guru Nanak Institutions Technical Campus, Hyderabad, India
| | - Jeetendra Kumar Gupta
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, Chaumuhan, Uttar Pradesh, India
| | - Kasturi Nagasree
- Department of Regulatory Affairs, Samskruthi College of Pharmacy Samskruti College of Pharmacy, Ghatkesar, Telangana, India
| | - R Lokeshvar
- Department of Pharmacology, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Saveetha Nagar, Thandalam, Chennai, India
| | - Selvaraja Elumalai
- Department of Quality Control, Ambiopharm Inc, Dittman Ct, Beach Island, South Carolina, 29842, USA
| | - P Dharani Prasad
- Department of Pharmacology, Mohan Babu University, MB School of Pharmaceutical Sciences, (Erstwhile, Sree Vidyaniketan College of Pharmacy), Tirupati, India
| | - Deepti Kolli
- Department of Chemistry, Koneru Lakshmaiah Education Foundation, Greenfields, Vaddeswaram, Guntur, Andhra Pradesh, India.
| |
Collapse
|
10
|
Baghel R, Maan K, Dhariwal S, Kumari M, Sharma A, Manda K, Trivedi R, Rana P. Mild Blast Exposure Dysregulates Metabolic Pathways and Correlation Networking as Evident from LC-MS-Based Plasma Profiling. Mol Neurobiol 2025; 62:3143-3166. [PMID: 39235645 DOI: 10.1007/s12035-024-04429-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 08/08/2024] [Indexed: 09/06/2024]
Abstract
Blast-induced trauma is emerging as a serious threat due to its wide pathophysiology where not only the brain but also a spectrum of organs is being affected. In the present study, we aim to identify the plasma-based metabolic dysregulations along with the associated temporal changes at 5-6 h, day 1 and day 7 post-injury in a preclinical animal model for blast exposure, through liquid chromatography-mass spectrometry (LC-MS). Using significantly advanced metabolomic and statistical bioinformatic platforms, we were able to elucidate better and unravel the complex networks of blast-induced neurotrauma (BINT) and its interlinked systemic effects. Significant changes were evident at 5-6 h with maximal changes at day 1. Temporal analysis also depicted progressive changes which continued till day 7. Significant associations of metabolic markers belonging to the class of amino acids, energy-related molecules, lipids, vitamin, hormone, phenolic acid, keto and histidine derivatives, nucleic acid molecules, uremic toxins, and uronic acids were observed. Also, the present study is the first of its kind where comprehensive, detailed pathway dysregulations of amino acid metabolism and biosynthesis, perturbed nucleotides, lipid peroxidation, and nucleic acid damage followed by correlation networking and multiomics networking were explored on preclinical animal models exposed to mild blast trauma. In addition, markers for systemic changes (renal dysfunction) were also observed. Global pathway predictions of unannotated peaks also presented important insights into BINT pathophysiology. Conclusively, the present study depicts important findings that might help underpin the biological mechanisms of blast-induced brain or systemic trauma.
Collapse
Affiliation(s)
- Ruchi Baghel
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Science (INMAS), DRDO, New Delhi, 110054, India
- Department of Health Research (DHR), IRCS Building, 2 FloorRed Cross Road, New Delhi, 110001, India
- Metabolomics Research Facility, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, S. K Mazumdar Road, Timarpur, New Delhi, 110054, India
| | - Kiran Maan
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Science (INMAS), DRDO, New Delhi, 110054, India
- Metabolomics Research Facility, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, S. K Mazumdar Road, Timarpur, New Delhi, 110054, India
| | - Seema Dhariwal
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Science (INMAS), DRDO, New Delhi, 110054, India
- Metabolomics Research Facility, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, S. K Mazumdar Road, Timarpur, New Delhi, 110054, India
| | - Megha Kumari
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Science (INMAS), DRDO, New Delhi, 110054, India
| | - Apoorva Sharma
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Science (INMAS), DRDO, New Delhi, 110054, India
- Metabolomics Research Facility, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, S. K Mazumdar Road, Timarpur, New Delhi, 110054, India
| | - Kailash Manda
- Department of Neurobehavioral Sciences, Institute of Nuclear Medicine and Allied Science (INMAS), DRDO, New Delhi, 110054, India
| | - Richa Trivedi
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Science (INMAS), DRDO, New Delhi, 110054, India
| | - Poonam Rana
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Science (INMAS), DRDO, New Delhi, 110054, India.
- Metabolomics Research Facility, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, S. K Mazumdar Road, Timarpur, New Delhi, 110054, India.
| |
Collapse
|
11
|
Gakpey MEL, Aidoo SA, Jumah TA, Hanson G, Msipa S, Mbaoji FN, Bukola O, Tjale PC, Sangare M, Tebourbi H, Awe OI. Targeting aldose reductase using natural African compounds as promising agents for managing diabetic complications. FRONTIERS IN BIOINFORMATICS 2025; 5:1499255. [PMID: 39996053 PMCID: PMC11848289 DOI: 10.3389/fbinf.2025.1499255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/17/2025] [Indexed: 02/26/2025] Open
Abstract
Background Diabetes remains a leading cause of morbidity and mortality due to various complications induced by hyperglycemia. Inhibiting Aldose Reductase (AR), an enzyme that converts glucose to sorbitol, has been studied to prevent long-term diabetic consequences. Unfortunately, drugs targeting AR have demonstrated toxicity, adverse reactions, and a lack of specificity. This study aims to explore African indigenous compounds with high specificity as potential AR inhibitors for pharmacological intervention. Methodology A total of 7,344 compounds from the AfroDB, EANPDB, and NANPDB databases were obtained and pre-filtered using the Lipinski rule of five to generate a compound library for virtual screening against the Aldose Reductase. The top 20 compounds with the highest binding affinity were selected. Subsequently, in silico analyses such as protein-ligand interaction, physicochemical and pharmacokinetic profiling (ADMET), and molecular dynamics simulation coupled with binding free energy calculations were performed to identify lead compounds with high binding affinity and low toxicity. Results Five natural compounds, namely, (+)-pipoxide, Zinc000095485961, Naamidine A, (-)-pipoxide, and 1,6-di-o-p-hydroxybenzoyl-beta-d-glucopyranoside, were identified as potential inhibitors of aldose reductase. Molecular docking results showed that these compounds exhibited binding energies ranging from -12.3 to -10.7 kcal/mol, which were better than the standard inhibitors (zopolrestat, epalrestat, IDD594, tolrestat, and sorbinil) used in this study. The ADMET and protein-ligand interaction results revealed that these compounds interacted with key inhibiting residues through hydrogen and hydrophobic interactions and demonstrated favorable pharmacological and low toxicity profiles. Prediction of biological activity highlighted Zinc000095485961 and 1,6-di-o-p-hydroxybenzoyl-beta-d-glucopyranoside as having significant inhibitory activity against aldose reductase. Molecular dynamics simulations and MM-PBSA analysis confirmed that the compounds bound to AR exhibited high stability and less conformational change to the AR-inhibitor complex. Conclusion This study highlighted the potential inhibitory activity of 5 compounds that belong to the African region: (+)-Pipoxide, Zinc000095485961, Naamidine A, (-)-Pipoxide, and 1,6-di-o-p-hydroxybenzoyl-beta-d-glucopyranoside. These molecules inhibiting the aldose reductase, the key enzyme of the polyol pathway, can be developed as therapeutic agents to manage diabetic complications. However, we recommend in vitro and in vivo studies to confirm our findings.
Collapse
Affiliation(s)
- Miriam E. L. Gakpey
- Department of Clinical Pathology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Shadrack A. Aidoo
- Department of Virology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Toheeb A. Jumah
- School of Collective Intelligence, University Mohammed VI Polytechnic, Rabat, Morocco
| | - George Hanson
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Siyabonga Msipa
- Department of Integrative Biomedical Science, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Florence N. Mbaoji
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, Enugu, Nigeria
| | - Omonijo Bukola
- Department of Medical Laboratory Science, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomosho, Oyo, Nigeria
| | - Palesa C. Tjale
- Department of Computational Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Mamadou Sangare
- African Center of Excellence in Bioinformatics (ACE-B), University of Science, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Hedia Tebourbi
- Pathophysiology, Food and Biomolecules Laboratory, Higher Institute of Biotechnology of Sidi Thabet, Sidi Thabet, Tunisia
| | - Olaitan I. Awe
- African Society for Bioinformatics and Computational Biology, Cape Town, South Africa
| |
Collapse
|
12
|
Pandey AK, Verma S, Singh R. Study of Pineapple Bioactive Compounds Targeting Aldose Reductase: A Natural Intervention for Diabetes Mellitus Pathologies. Mol Biotechnol 2025:10.1007/s12033-025-01380-1. [PMID: 39891846 DOI: 10.1007/s12033-025-01380-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/10/2025] [Indexed: 02/03/2025]
Abstract
Aldose reductase is a reduced monomeric enzyme that utilizes NADPH as a cofactor to mediate the glucose reduction to sorbitol in the polyol pathway. Overexpression of aldose reductase has been observed to mediate pathologies associated with diabetes mellitus. Inhibition of aldose reductase thus seems promising to deal with these pathologies. Pineapple and its extract have been identified for its anti-diabetic effect due to the presence of effective bioactive agents. In the present study, the major bioactive compounds of pineapple have been studied for their potential to structurally inhibit aldose reductase. The ADMET analysis of lead bioactive compounds including myrcene, palmitic acid, limonene, n-decanal, beta-carophyllene, 1-cyclohexane-1-caboxaldehyde, and α-farnesene showed most of the compounds were non-toxic and have druglike properties with LD50 values of greater than 2000 mg/kg. Molecular docking of these compounds at the substrate binding site of the aldose reductase-NADPH complex disclosed effective binding with binding energy values of - 5.025 to - 8.003 kcal/mol. α-farnesene, known for its antibacterial, antiviral, and anti-inflammatory properties gave the highest binding energy of - 8.003 kcal/mol. The molecular dynamic simulation studies of α-farnesene-aldose reductase-NADPH ternary complex, aldose reductase-NADPH binary complex, and apo-aldose reductase revealed similar RMSD values with respect to time during the simulation trajectory indicating stable interaction of the compound with the enzyme. DFT analysis showed high reactivity of α-farnesene which favours its utilization as a drug for specific target protein. Therefore, this study provides an efficient natural aldose reductase inhibitor α-farnesene that can be further explored for its potential to develop an effective natural drug to treat diabetes.
Collapse
Affiliation(s)
- Anand Kumar Pandey
- Department of Biotechnology Engineering, Institute of Engineering and Technology, Bundelkhand University, Jhansi, Uttar Pradesh, 284128, India.
| | - Shalja Verma
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand, 247667, India
| | - Rupanjali Singh
- Department of Biotechnology Engineering, Institute of Engineering and Technology, Bundelkhand University, Jhansi, Uttar Pradesh, 284128, India
| |
Collapse
|
13
|
Roney M, Issahaku AR, Uddin MN, Wilhelm A, Aluwi MFFM. Exploration of leads from bis-indole based triazine derivatives targeting human aldose reductase in diabetic type 2: in-silico approaches. 3 Biotech 2025; 15:5. [PMID: 39676892 PMCID: PMC11635081 DOI: 10.1007/s13205-024-04178-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/26/2024] [Indexed: 12/17/2024] Open
Abstract
Diabetes mellitus (DM) poses a major global healthcare challenge, highlighting the need for new treatments beyond current options. Currently available drugs have side effects including weight gain, nausea, vomiting, diarrhea, insulin resistance etc. Therefore, given the benefits of indole derivatives in diabetes and the lack of computational studies on bis-indole-based triazine derivatives with aldose reductase (AR), this study employs in-silico analysis to explore their potential as type-2 diabetes treatments. Based on the Differential Expression analysis, the human aldose reductase (HAR) encoding gene AKR1B1 showed overexpression in GSE30122 diabetes patients (Log2FC = 0.62, P < 0.01). Moreover, the compounds 2-((5,6-di(1H-indol-3-yl)-1,2,4-triazin-3-yl)thio)-1-(3-hydroxy-5-methylphenyl)ethan-1-one (4) and 2-((5,6-di(1H-indol-3-yl)-1,2,4-triazin-3-yl)thio)-1-(4-nitrophenyl)ethan-1-one (8) were identified as leading candidates, showing binding energies of - 62.12, - 81.73 kcal/mol and - 57.19, - 85.97 kcal/mol, respectively. Docking, MM/GBSA screening, molecular dynamics (MD) simulations, PCA, and post-MM/GBSA analysis confirmed their stability and favorable binding compared to the apo protein and control. Further in-vitro, in-vivo, and clinical studies are required to validate their therapeutic potential.
Collapse
Affiliation(s)
- Miah Roney
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang Al-Sultan Abdullah, Lebuhraya Tun Razak, Gambang, 26300 Kuantan, Pahang Malaysia
- Centre for Bio-Aromatic Research, Universiti Malaysia Pahang Al-Sultan Abdullah, Lebuhraya Tun Razak, Gambang, 26300 Kuantan, Pahang Malaysia
| | - Abdul Rashid Issahaku
- Department of Chemistry, University of the Free State, 205 Nelson Mandela Avenue, Bloemfontein, 9301 South Africa
| | - Md. Nazim Uddin
- Institute of Food Science and Technology, Bangladesh Council of Scientific and Industrial Research, Dhaka, 1205 Bangladesh
| | - Anke Wilhelm
- Department of Chemistry, University of the Free State, 205 Nelson Mandela Avenue, Bloemfontein, 9301 South Africa
| | - Mohd Fadhlizil Fasihi Mohd Aluwi
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang Al-Sultan Abdullah, Lebuhraya Tun Razak, Gambang, 26300 Kuantan, Pahang Malaysia
- Centre for Bio-Aromatic Research, Universiti Malaysia Pahang Al-Sultan Abdullah, Lebuhraya Tun Razak, Gambang, 26300 Kuantan, Pahang Malaysia
| |
Collapse
|
14
|
Sharma S, Choudhary M, Sharma O, Injeti E, Mittal A. Mechanistic insights into antidiabetic potential of Ficus viren against multi organ specific diabetic targets: molecular docking, MDS, MM-GBSA analysis. Comput Biol Chem 2024; 113:108185. [PMID: 39217892 DOI: 10.1016/j.compbiolchem.2024.108185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/19/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Ficus viren has been traditionally used to treat diabetes, and its extract inhibits carbohydrate/lipid metabolism and possesses anti-hyperglycemic potential. However, there is conflicting investigation related to F. viren extract effect on carbohydrate metabolism. Thus, bioactive and mechanism behind its antidiabetic potential is still scanty. This study explored F. viren's anti-diabetic property by identifying potential phytoconstituents and mechanism. A sequential in-silico approach was used i.e., druglikeness, molecular docking, post-docking MM-GBSA, ADMET studies, molecular dynamic simulation (MDS), and post-MDS MM-GBSA. We screened ∼32 phytoconstituents and twelve potential organ-specific diabetic targets (O.S.D.Ts i.e., IR, DPP-4, ppar-γ, ppar-α, ppar-δ, GLP-1R, SIRT-1, AMPK, GSK-3β, RAGE, and AR). Drug likeness study identified 18 druggable candidates among 32 phytoconstituents. K3A, quercetin, scutellarein, sorbifolin, and vogeline J identified as potential ligands from druggable ligands, using IR as the standard target. Subsequently, potential ligands docked with remaining O.S.D.Ts. and data showed that K3A binds strongly with AMPK, ppar-δ, DPP-4, and GSK-3β, while scutellarein binds with AR and ppar-α. Sorbifolin, quercetin, and vogeline J binds with ppar-α, ppar-γ, and RAGE, respectively. Post-docking MM-GBSA data (∆GBind) also depicted potential ligand's strong binding affinities with their corresponding targets. Thereafter, simulation data revealed that only scutellarein and sorbifolin showed dynamic stability with their respective targets, i.e., AR/ppar-α and ppar-α, respectively. Interestingly, post-MDS MM-GBSA revealed that only scutellarein exhibited strong ∆GBind of -55.08 kcal/mol and -75.48 kcal/mol with AR and ppar-α, respectively. Though, collective computational analysis supports antidiabetic potential of F. viren through AR and ppar-α modulation by scutellarein.
Collapse
Affiliation(s)
- Sachin Sharma
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra 136119, India
| | - Manjusha Choudhary
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra 136119, India
| | - Onkar Sharma
- Skeletal Muscle Lab, IIHS, Kurukshetra University, Kurukshetra, Haryana 136119, India
| | - Elisha Injeti
- Department of Pharmaceutical Sciences, Cedarville University, Cedarville, OH 45314, USA
| | - Ashwani Mittal
- Skeletal Muscle Lab, IIHS, Kurukshetra University, Kurukshetra, Haryana 136119, India.
| |
Collapse
|
15
|
Evans AJ, Li YL. Remodeling of the Intracardiac Ganglia During the Development of Cardiovascular Autonomic Dysfunction in Type 2 Diabetes: Molecular Mechanisms and Therapeutics. Int J Mol Sci 2024; 25:12464. [PMID: 39596529 PMCID: PMC11594459 DOI: 10.3390/ijms252212464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is one of the most significant health issues worldwide, with associated healthcare costs estimated to surpass USD 1054 billion by 2045. The leading cause of death in T2DM patients is the development of cardiovascular disease (CVD). In the early stages of T2DM, patients develop cardiovascular autonomic dysfunction due to the withdrawal of cardiac parasympathetic activity. Diminished cardiac parasympathetic tone can lead to cardiac arrhythmia-related sudden cardiac death, which accounts for 50% of CVD-related deaths in T2DM patients. Regulation of cardiovascular parasympathetic activity is integrated by neural circuitry at multiple levels including afferent, central, and efferent components. Efferent control of cardiac parasympathetic autonomic tone is mediated through the activity of preganglionic parasympathetic neurons located in the cardiac extensions of the vagus nerve that signals to postganglionic parasympathetic neurons located in the intracardiac ganglia (ICG) on the heart. Postganglionic parasympathetic neurons exert local control on the heart, independent of higher brain centers, through the release of neurotransmitters, such as acetylcholine. Structural and functional alterations in cardiac parasympathetic postganglionic neurons contribute to the withdrawal of cardiac parasympathetic tone, resulting in arrhythmogenesis and sudden cardiac death. This review provides an overview of the remodeling of parasympathetic postganglionic neurons in the ICG, and potential mechanisms contributing to the withdrawal of cardiac parasympathetic tone, ventricular arrhythmogenesis, and sudden cardiac death in T2DM. Improving cardiac parasympathetic tone could be a therapeutic avenue to reduce malignant ventricular arrhythmia and sudden cardiac death, increasing both the lifespan and improving quality of life of T2DM patients.
Collapse
Affiliation(s)
- Anthony J. Evans
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
16
|
Klochkov V, Chan CM, Lin WW. Methylglyoxal: A Key Factor for Diabetic Retinopathy and Its Effects on Retinal Damage. Biomedicines 2024; 12:2512. [PMID: 39595078 PMCID: PMC11592103 DOI: 10.3390/biomedicines12112512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Diabetic retinopathy is the most common retinal vascular disease, affecting the retina's blood vessels and causing chronic inflammation, oxidative stress, and, ultimately, vision loss. Diabetes-induced elevated glucose levels increase glycolysis, the main methylglyoxal (MGO) formation pathway. MGO is a highly reactive dicarbonyl and the most rapid glycation compound to form endogenous advanced glycation end products (AGEs). MGO can act both intra- and extracellularly by glycating molecules and activating the receptor for AGEs (RAGE) pathway. Conclusions: This review summarizes the sources of MGO formation and its actions on various cell pathways in retinal cells such as oxidative stress, glycation, autophagy, ER stress, and mitochondrial dysfunction. Finally, the detoxification of MGO by glyoxalases is discussed.
Collapse
Affiliation(s)
- Vladlen Klochkov
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Ophthalmology, Cardinal Tien Hospital, New Taipei City 23148, Taiwan
| | - Chi-Ming Chan
- Department of Ophthalmology, Cardinal Tien Hospital, New Taipei City 23148, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| | - Wan-Wan Lin
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 100233, Taiwan
| |
Collapse
|
17
|
Kattar A, Vivero-Lopez M, Concheiro A, Mudakavi R, Chauhan A, Alvarez-Lorenzo C. Oleogels for the ocular delivery of epalrestat: formulation, in vitro, in ovo, ex vivo and in vivo evaluation. Drug Deliv Transl Res 2024; 14:3291-3308. [PMID: 38780858 PMCID: PMC11445291 DOI: 10.1007/s13346-024-01560-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2024] [Indexed: 05/25/2024]
Abstract
The ocular administration of lipophilic and labile drugs such as epalrestat, an aldose reductase inhibitor with potential for diabetic retinopathy treatment, demands the development of topical delivery systems capable of providing sufficient ocular bioavailability. The aim of this work was to develop non-aqueous oleogels based on soybean oil and gelators from natural and sustainable sources (ethyl cellulose, beeswax and cocoa butter) and to assess their reproducibility, safety and efficiency in epalrestat release and permeation both ex vivo and in vivo. Binary combinations of gelators at 10% w/w resulted in solid oleogels (oleorods), while single gelator oleogels at 5% w/w remained liquid at room temperature, with most of the oleogels displaying shear thinning behavior. The oleorods released up to 4 µg epalrestat per mg of oleorod in a sustained or burst pattern depending on the gelator (approx. 10% dose in 24 h). The HET-CAM assay indicated that oleogel formulations did not induce ocular irritation and were safe for topical ocular administration. Corneal and scleral ex vivo assays evidenced the permeation of epalrestat from the oleorods up to 4 and 2.5 µg/cm2 after six hours, respectively. Finally, the capacity of the developed oleogels to sustain release and provide significant amounts of epalrestat to the ocular tissues was demonstrated in vivo against aqueous-based niosomes and micelles formulations loaded with the same drug concentration. Overall, the gathered information provides valuable insights into the development of oleogels for ocular drug delivery, emphasizing their safety and controlled release capabilities, which have implications for the treatment of diabetic neuropathy and other ocular conditions.
Collapse
Affiliation(s)
- Axel Kattar
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Maria Vivero-Lopez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Rajeev Mudakavi
- Department of Chemical Engineering, Colorado School of Mines, Golden, CO, 80401, USA
| | - Anuj Chauhan
- Department of Chemical Engineering, Colorado School of Mines, Golden, CO, 80401, USA
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain.
| |
Collapse
|
18
|
Gupta S, Jha S, Rani S, Arora P, Kumar S. Medicinal Perspective of 2,4-Thiazolidinediones Derivatives: An Insight into Recent Advancements. ChemistryOpen 2024; 13:e202400147. [PMID: 39246226 PMCID: PMC11564877 DOI: 10.1002/open.202400147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/25/2024] [Indexed: 09/10/2024] Open
Abstract
2,4-Thiazolidinedione derivatives represent nitrogen-containing heterocyclic compounds utilized in type 2 diabetes mellitus management. Recent advances in medicinal chemistry have unveiled diverse therapeutic potentials and structural modifications of these derivatives. This review delves into novel TZD derivatives, encompassing their synthesis, structure-activity relationships, and pharmacokinetic profiles. Various therapeutic potentials of TZDs are explored, including anticancer, antimicrobial, anti-inflammatory, antioxidant, anticonvulsant, antihyperlipidemic, anticorrosive, and antitubercular activities. Additionally, it addresses mitigating side effects associated with marketed TZD derivatives such as weight gain, oedema, fractures, and congestive heart failure in type 2 diabetes mellitus management. The review elaborates on in vivo, in vitro, and ex vivo studies supporting different biological activities, alongside predicting ADME and drug-likeness properties of TZDs. Computational studies are also integrated to elucidate binding modes and affinities of novel TZD derivatives. Furthermore, a plethora of novel TZD derivatives with varied and enhanced therapeutic potentials are presented, warranting further evaluation of their biological activities.
Collapse
Affiliation(s)
- Sneha Gupta
- School of Pharmaceutical SciencesLovely Professional UniversityJalandhar-Delhi G.T. RoadPhagwaraPunjab144411India
| | - Sumeet Jha
- School of Pharmaceutical SciencesLovely Professional UniversityJalandhar-Delhi G.T. RoadPhagwaraPunjab144411India
| | - Supriya Rani
- School of Pharmaceutical SciencesLovely Professional UniversityJalandhar-Delhi G.T. RoadPhagwaraPunjab144411India
| | - Pinky Arora
- School of bioengineering and biosciencesLovely Professional UniversityJalandhar-Delhi G.T. RoadPhagwaraPunjab144411India
| | - Shubham Kumar
- School of Pharmaceutical SciencesLovely Professional UniversityJalandhar-Delhi G.T. RoadPhagwaraPunjab144411India
| |
Collapse
|
19
|
Ahmad S, Ahmad MFA, Khan S, Alouffi S, Khan M, Prakash C, Khan MWA, Ansari IA. Exploring aldose reductase inhibitors as promising therapeutic targets for diabetes-linked disabilities. Int J Biol Macromol 2024; 280:135761. [PMID: 39306154 DOI: 10.1016/j.ijbiomac.2024.135761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024]
Abstract
Diabetes mellitus significantly increases mortality and morbidity rates due to complications like neuropathy and nephropathy. It also leads to retinopathy and cataract formation, which is a leading cause of vision disability. The polyol pathway emerges as a promising therapeutic target among the various pathways associated with diabetic complications. This review focuses on the development of natural and synthetic aldose reductase inhibitors (ARIs), along with recent discoveries in diabetic complication treatment. AR, pivotal in the polyol pathway converting glucose to sorbitol, plays a key role in secondary diabetes complications' pathophysiology. Understanding AR's function and structure lays the groundwork for improving ARIs to mitigate diabetic complications. New developments in ARIs open up exciting possibilities for treating diabetes-related complications. However, it is still challenging to get preclinical successes to clinical effectiveness because of things like differences in how the disease starts, drug specificity, and the complexity of the AR's structure. Addressing these challenges is crucial for developing targeted and efficient ARIs. Continued research into AR's structural features and specific ARIs is essential. Overcoming these challenges could revolutionize diabetic complication treatment, enhance patient outcomes, and reduce the global burden of diabetes-related mortality and morbidity.
Collapse
Affiliation(s)
- Saheem Ahmad
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, 2440, Saudi Arabia.
| | | | - Saif Khan
- Department of Basic Dental and Medical Sciences, College of Dentistry, University of Hail, Saudi Arabia
| | - Sultan Alouffi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, 2440, Saudi Arabia
| | - Mahvish Khan
- Department of Biology, College of Science, University of Hail, 2440, Saudi Arabia
| | - Chander Prakash
- University Centre for Research and Development, Chandigarh University, Mohali, Punjab, India
| | - Mohd Wajid Ali Khan
- Department of Chemistry, College of Science, University of Hail, 2440, Saudi Arabia; Medical and Diagnostic Research Center, University of Ha'il, Ha'il-55473, Saudi Arabia
| | - Irfan Ahmad Ansari
- Department of Biology, College of Science, University of Hail, 2440, Saudi Arabia.
| |
Collapse
|
20
|
Türkeş C. Aldose reductase with quinolone antibiotics interaction: In vitro and in silico approach of its relationship with diabetic complications. Arch Biochem Biophys 2024; 761:110161. [PMID: 39313142 DOI: 10.1016/j.abb.2024.110161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
Aldose reductase (AR, EC1.1.1.21), a member of the aldo-keto reductase family, is critically implicated in the pathogenesis of chronic complications associated with diabetes mellitus, including neuropathy, nephropathy, and retinopathy. Hyperglycemia-induced AR overactivity results in intracellular sorbitol accumulation, NADPH depletion, and oxidative stress. Consequently, AR is recognized as a key mediator of oxidative and inflammatory signaling pathways involved in diverse human pathologies such as cardiovascular diseases, inflammatory disorders, and cancer. This has sparked renewed interest in developing novel AR inhibitors (ARIs) with enhanced therapeutic profiles. In this study, we evaluated the inhibitory potential of five quinolone antibiotics-gatifloxacin, lomefloxacin, nalidixic acid, norfloxacin, and sparfloxacin-as ARIs relevant to various physiological and pathological conditions. Through comprehensive in vitro and in silico analyses, we explored these antibiotics' binding interactions and affinities within the AR active site. Our findings reveal that these quinolones moderately inhibit AR at micromolar concentrations, with inhibition constants (KIs) ranging from 1.03 ± 0.13 μM to 4.12 ± 0.51 μM, compared to the reference drug epalrestat (KI of 0.85 ± 0.06 μM). The combined in vitro and in silico results underscore significant interactions between these drugs and AR, suggesting their potential as therapeutic agents against the aforementioned pathological conditions. Furthermore, these insights will aid in optimizing clinical dosing regimens and mitigating unexpected drug-drug interactions when these antibiotics are co-administered with other treatments.
Collapse
Affiliation(s)
- Cüneyt Türkeş
- Department of Biochemistry, Faculty of Pharmacy, Erzincan Binali Yıldırım University, Erzincan, 24002, Turkey.
| |
Collapse
|
21
|
Fan R, Kong J, Zhang J, Zhu L. Exercise as a therapeutic approach to alleviate diabetic kidney disease: mechanisms, clinical evidence and potential exercise prescriptions. Front Med (Lausanne) 2024; 11:1471642. [PMID: 39526249 PMCID: PMC11543430 DOI: 10.3389/fmed.2024.1471642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Diabetic kidney disease (DKD) is a global and severe complication that imposes a significant burden on individual health, families, and society. Currently, the main treatment approaches for DKD include medication, blood glucose control, protein-restricted diet, and blood pressure management, all of which have certain limitations. Exercise, as a non-pharmacological intervention, has attracted increasing attention. This review introduces the mechanisms and clinical evidence of exercise on DKD, and proposes potential exercise prescriptions. Exercise can improve blood glucose stability related to DKD and the renin-angiotensin-aldosterone system (RAAS), reduce renal oxidative stress and inflammation, enhance the crosstalk between muscle and kidneys, and improve endothelial cell function. These mechanisms contribute to the comprehensive improvement of DKD. Compared to traditional treatment methods, exercise has several advantages, including safety, effectiveness, and no significant side effects. It can be used as an adjunct therapy to medication, blood glucose control, protein-restricted diet, and blood pressure management. Despite the evident benefits of exercise in DKD management, there is still a lack of large-scale, long-term randomized controlled trials to provide more evidence and develop exercise guidelines for DKD. Healthcare professionals should actively encourage exercise in DKD patients and develop personalized exercise plans based on individual circumstances.
Collapse
Affiliation(s)
| | | | | | - Lei Zhu
- College of Sports Science, Qufu Normal University, Qufu, China
| |
Collapse
|
22
|
Yasir M, Park J, Han ET, Han JH, Park WS, Chun W. Investigating the Inhibitory Potential of Flavonoids against Aldose Reductase: Insights from Molecular Docking, Dynamics Simulations, and gmx_MMPBSA Analysis. Curr Issues Mol Biol 2024; 46:11503-11518. [PMID: 39451563 PMCID: PMC11506312 DOI: 10.3390/cimb46100683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/26/2024] Open
Abstract
Diabetes mellitus (DM) is a complex metabolic disorder characterized by chronic hyperglycemia, with aldose reductase playing a critical role in the pathophysiology of diabetic complications. This study aimed to investigate the efficacy of flavonoid compounds as potential aldose reductase inhibitors using a combination of molecular docking and molecular dynamics (MD) simulations. The three-dimensional structures of representative flavonoid compounds were obtained from PubChem, minimized, and docked against aldose reductase using Discovery Studio's CDocker module. The top 10 compounds Daidzein, Quercetin, Kaempferol, Butin, Genistein, Sterubin, Baicalein, Pulchellidin, Wogonin, and Biochanin_A were selected based on their lowest docking energy values for further analysis. Subsequent MD simulations over 100 ns revealed that Daidzein and Quercetin maintained the highest stability, forming multiple conventional hydrogen bonds and strong hydrophobic interactions, consistent with their favorable interaction energies and stable RMSD values. Comparative analysis of hydrogen bond interactions and RMSD profiles underscored the ligand stability. MMPBSA analysis further confirmed the significant binding affinities of Daidzein and Quercetin, highlighting their potential as aldose reductase inhibitors. This study highlights the potential of flavonoids as aldose reductase inhibitors, offering insights into their binding interactions and stability, which could contribute to developing novel therapeutics for DM complications.
Collapse
Affiliation(s)
- Muhammad Yasir
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (M.Y.); (J.P.)
| | - Jinyoung Park
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (M.Y.); (J.P.)
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (E.-T.H.); (J.-H.H.)
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (E.-T.H.); (J.-H.H.)
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea;
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea; (M.Y.); (J.P.)
| |
Collapse
|
23
|
Frusciante L, Geminiani M, Shabab B, Olmastroni T, Scavello G, Rossi M, Mastroeni P, Nyong'a CN, Salvini L, Lamponi S, Parisi ML, Sinicropi A, Costa L, Spiga O, Trezza A, Santucci A. Exploring the Antioxidant and Anti-Inflammatory Potential of Saffron ( Crocus sativus) Tepals Extract within the Circular Bioeconomy. Antioxidants (Basel) 2024; 13:1082. [PMID: 39334741 PMCID: PMC11428576 DOI: 10.3390/antiox13091082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Repurposing saffron (Crocus sativus) waste presents a sustainable strategy for generating high-value products within the bioeconomy framework. Typically, flower components are discarded after stigma harvest, resulting in significant waste-350 kg of tepals per kilogram of stigmas. This research employed a comprehensive approach, integrating bioactivity studies (in vitro and in silico) with Life Cycle Assessment (LCA) evaluations, to extract and assess bioactive compounds from C. sativus tepals sourced in Tuscany, Italy. Phytochemical characterization using UPLC-MS/MS revealed a high abundance and variety of flavonoids in the hydro-ethanolic extract (CST). The antioxidant capacity was validated through various assays, and the ability to mitigate H2O2-induced oxidative stress and enhance fermentation was demonstrated in Saccharomyces cerevisiae. This study reports that C. sativus tepals extract reduces oxidative stress and boosts ethanol fermentation in yeast, paving the way for applications in the food and biofuels sectors. Further validation in RAW 264.7 macrophages confirmed CST's significant anti-inflammatory effects, indicating its potential for pharmaceutical, cosmeceutical, and nutraceutical applications. In silico studies identified potential targets involved in antioxidant and anti-inflammatory processes, shedding light on possible interaction mechanisms with Kaempferol 3-O-sophoroside (KOS-3), the predominant compound in the extract. The integration of LCA studies highlighted the environmental benefits of this approach. Overall, this research underscores the value of using waste-derived extracts through "green" methodologies, offering a model that may provide significant advantages for further evaluations compared to traditional methodologies and supporting the circular bioeconomy.
Collapse
Affiliation(s)
- Luisa Frusciante
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Michela Geminiani
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
- SienabioACTIVE, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Behnaz Shabab
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Tommaso Olmastroni
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Giorgia Scavello
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Martina Rossi
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Pierfrancesco Mastroeni
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Collins Nyaberi Nyong'a
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Laura Salvini
- Fondazione Toscana Life Sciences, Strada del Petriccio e Belriguardo, 53100 Siena, Italy
| | - Stefania Lamponi
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
- SienabioACTIVE, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Maria Laura Parisi
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
- LifeCARES, Via Emilio Vezzosi 15, 52100 Arezzo, Italy
| | - Adalgisa Sinicropi
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
- LifeCARES, Via Emilio Vezzosi 15, 52100 Arezzo, Italy
| | - Lorenzo Costa
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Ottavia Spiga
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
- ARTES 4.0, Viale Rinaldo Piaggio, 34, 56025 Pontedera, Italy
| | - Alfonso Trezza
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Annalisa Santucci
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
- SienabioACTIVE, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
- ARTES 4.0, Viale Rinaldo Piaggio, 34, 56025 Pontedera, Italy
| |
Collapse
|
24
|
Peng J, Abdulla R, Liu X, He F, Xin X, Aisa HA. Polyphenol-Rich Extract of Apocynum venetum L. Leaves Protects Human Retinal Pigment Epithelial Cells against High Glucose-Induced Damage through Polyol Pathway and Autophagy. Nutrients 2024; 16:2944. [PMID: 39275261 PMCID: PMC11397065 DOI: 10.3390/nu16172944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Diabetic retinopathy (DR) is a specific microvascular problem of diabetes, which is mainly caused by hyperglycemia and may lead to rapid vision loss. Dietary polyphenols have been reported to decrease the risk of DR. Apocynum venetum L. leaves are rich in polyphenolic compounds and are popular worldwide for their health benefits as a national tea drink. Building on previous findings of antioxidant activity and aldose reductase inhibition of A. venetum, this study investigated the chemical composition of polyphenol-rich extract of A. venetum leaves (AVL) and its protective mechanism on ARPE-19 cells in hyperglycemia. Ninety-three compounds were identified from AVL by LC-MS/MS, including sixty-eight flavonoids, twenty-one organic acids, and four coumarins. AVL regulated the polyol pathway by decreasing the expression of aldose reductase and the content of sorbitol, enhancing the Na+K+-ATPase activity, and weakening intracellular oxidative stress effectively; it also could regulate the expression of autophagy-related proteins via the AMPK/mTOR/ULK1 signaling pathway to maintain intracellular homeostasis. AVL could restore the polyol pathway, inhibit oxidative stress, and maintain intracellular autophagy to protect cellular morphology and improve DR. The study reveals the phytochemical composition and protective mechanisms of AVL against DR, which could be developed as a functional food and/or candidate pharmaceutical, aiming for retina protection in diabetic retinopathy.
Collapse
Affiliation(s)
- Jun Peng
- The State Key Laboratory Basis Xinjiang Indigenous Medicinal Plant Resource, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Rahima Abdulla
- The State Key Laboratory Basis Xinjiang Indigenous Medicinal Plant Resource, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
| | - Xiaoyan Liu
- The State Key Laboratory Basis Xinjiang Indigenous Medicinal Plant Resource, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Fei He
- The State Key Laboratory Basis Xinjiang Indigenous Medicinal Plant Resource, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
| | - Xuelei Xin
- The State Key Laboratory Basis Xinjiang Indigenous Medicinal Plant Resource, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
| | - Haji Akber Aisa
- The State Key Laboratory Basis Xinjiang Indigenous Medicinal Plant Resource, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
| |
Collapse
|
25
|
Khan LA, Khan MS, Ambrosy AP, Greene SJ. Selective aldose reductase inhibition as a treatment for diabetic cardiomyopathy: summary of the ARISE-HF trial. Heart Fail Rev 2024; 29:1157-1160. [PMID: 39052147 DOI: 10.1007/s10741-024-10427-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Affiliation(s)
- Laibah Arshad Khan
- Department of Medicine, King Edward Medical University, Lahore, 54000, Pakistan
| | - Muhammad Shahzeb Khan
- Division of Cardiology, Duke University School of Medicine, Duke University Medical Center, Durham, NC, 27705, USA
- Duke Clinical Research Institute, 300 West Morgan Street, Durham, NC, 27701, USA
| | - Andrew P Ambrosy
- Department of Cardiology, Kaiser Permanente San Francisco Medical Center, San Francisco, CA, USA
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Stephen J Greene
- Division of Cardiology, Duke University School of Medicine, Duke University Medical Center, Durham, NC, 27705, USA.
- Duke Clinical Research Institute, 300 West Morgan Street, Durham, NC, 27701, USA.
| |
Collapse
|
26
|
Yapislar H, Gurler EB. Management of Microcomplications of Diabetes Mellitus: Challenges, Current Trends, and Future Perspectives in Treatment. Biomedicines 2024; 12:1958. [PMID: 39335472 PMCID: PMC11429415 DOI: 10.3390/biomedicines12091958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/30/2024] Open
Abstract
Diabetes mellitus is a chronic metabolic disorder characterized by high blood sugar levels, which can lead to severe health issues if not managed effectively. Recent statistics indicate a significant global impact, with 463 million adults diagnosed worldwide and this projected to rise to 700 million by 2045. Type 1 diabetes is an autoimmune disorder where the immune system attacks pancreatic beta cells, reducing insulin production. Type 2 diabetes is primarily due to insulin resistance. Both types of diabetes are linked to severe microvascular and macrovascular complications if unmanaged. Microvascular complications, such as diabetic retinopathy, nephropathy, and neuropathy, result from damage to small blood vessels and can lead to organ and tissue dysfunction. Chronic hyperglycemia plays a central role in the onset of these complications, with prolonged high blood sugar levels causing extensive vascular damage. The emerging treatments and current research focus on various aspects, from insulin resistance to the intricate cellular damage induced by glucose toxicity. Understanding and intervening in these pathways are critical for developing effective treatments and managing diabetes long term. Furthermore, ongoing health initiatives, such as increasing awareness, encouraging early detection, and improving treatments, are in place to manage diabetes globally and mitigate its impact on health and society. These initiatives are a testament to the collective effort to combat this global health challenge.
Collapse
Affiliation(s)
- Hande Yapislar
- Department of Physiology, Faculty of Medicine, Acibadem University, 34752 Istanbul, Türkiye
| | - Esra Bihter Gurler
- Department of Basic Sciences, Faculty of Dentistry, Istanbul Galata University, 34430 Istanbul, Türkiye
| |
Collapse
|
27
|
Lei S, Liu C, Zheng TX, Fu W, Huang MZ. The relationship of redox signaling with the risk for atherosclerosis. Front Pharmacol 2024; 15:1430293. [PMID: 39148537 PMCID: PMC11324460 DOI: 10.3389/fphar.2024.1430293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/09/2024] [Indexed: 08/17/2024] Open
Abstract
Oxidative balance plays a pivotal role in physiological homeostasis, and many diseases, particularly age-related conditions, are closely associated with oxidative imbalance. While the strategic role of oxidative regulation in various diseases is well-established, the specific involvement of oxidative stress in atherosclerosis remains elusive. Atherosclerosis is a chronic inflammatory disorder characterized by plaque formation within the arteries. Alterations in the oxidative status of vascular tissues are linked to the onset, progression, and outcome of atherosclerosis. This review examines the role of redox signaling in atherosclerosis, including its impact on risk factors such as dyslipidemia, hyperglycemia, inflammation, and unhealthy lifestyle, along with dysregulation, vascular homeostasis, immune system interaction, and therapeutic considerations. Understanding redox signal transduction and the regulation of redox signaling will offer valuable insights into the pathogenesis of atherosclerosis and guide the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Sujuan Lei
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Chen Liu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Tian-Xiang Zheng
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary Surgery), Chongqing, Sichuan, China
| | - Wenguang Fu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary Surgery), Chongqing, Sichuan, China
| | - Mei-Zhou Huang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary Surgery), Chongqing, Sichuan, China
| |
Collapse
|
28
|
Januzzi JL, Butler J, Del Prato S, Ezekowitz JA, Ibrahim NE, Lam CSP, Lewis GD, Marwick TH, Perfetti R, Rosenstock J, Solomon SD, Tang WHW, Zannad F. Randomized Trial of a Selective Aldose Reductase Inhibitor in Patients With Diabetic Cardiomyopathy. J Am Coll Cardiol 2024; 84:137-148. [PMID: 38597864 DOI: 10.1016/j.jacc.2024.03.380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Progression to symptomatic heart failure is a complication of type 2 diabetes; heart failure onset in this setting is commonly preceded by deterioration in exercise capacity. OBJECTIVES This study sought to determine whether AT-001, a highly selective aldose reductase inhibitor, can stabilize exercise capacity among individuals with diabetic cardiomyopathy (DbCM) and reduced peak oxygen uptake (Vo2). METHODS A total of 691 individuals with DbCM meeting inclusion and exclusion criteria were randomized to receive placebo or ascending doses of AT-001 twice daily. Stratification at inclusion included region of enrollment, cardiopulmonary exercise test results, and use of sodium-glucose cotransporter 2 inhibitors or glucagon-like peptide-1 receptor agonists. The primary endpoint was proportional change in peak Vo2 from baseline to 15 months. Subgroup analyses included measures of disease severity and stratification variables. RESULTS The mean age was 67.5 ± 7.2 years, and 50.4% of participants were women. By 15 months, peak Vo2 fell in the placebo-treated patients by -0.31 mL/kg/min (P = 0.005 compared to baseline), whereas in those receiving high-dose AT-001, peak Vo2 fell by -0.01 mL/kg/min (P = 0.21); the difference in peak Vo2 between placebo and high-dose AT-001 was 0.30 (P = 0.19). In prespecified subgroup analyses among those not receiving sodium-glucose cotransporter 2 inhibitors or glucagon-like peptide-1 receptor agonists at baseline, the difference between peak Vo2 in placebo vs high-dose AT-001 at 15 months was 0.62 mL/kg/min (P = 0.04; interaction P = 0.10). CONCLUSIONS Among individuals with DbCM and impaired exercise capacity, treatment with AT-001 for 15 months did not result in significantly better exercise capacity compared with placebo. (Safety and Efficacy of AT-001 in Patients With Diabetic Cardiomyopathy [ARISE-HF]; NCT04083339).
Collapse
Affiliation(s)
- James L Januzzi
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA; Baim Institute for Clinical Research, Boston, Massachusetts, USA.
| | - Javed Butler
- University of Mississippi Medical Center, Jackson, Mississippi, USA; Baylor Scott and White Institute, Dallas, Texas, USA
| | - Stefano Del Prato
- Interdisciplinary Center "Health Sciences," Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Justin A Ezekowitz
- Division of Cardiology, University of Alberta, Edmonton, Alberta, Canada
| | - Nasrien E Ibrahim
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Carolyn S P Lam
- Baim Institute for Clinical Research, Boston, Massachusetts, USA; National Heart Centre Singapore and Duke-National University of Singapore, Singapore
| | - Gregory D Lewis
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas H Marwick
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | - Julio Rosenstock
- Velocity Clinical Research Center at Medical City, Dallas, Texas, USA
| | - Scott D Solomon
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - W H Wilson Tang
- Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Faiez Zannad
- Université de Lorraine, Inserm CIC and CHRU, Nancy, France
| |
Collapse
|
29
|
Yamaguchi H, Matsumura T, Sugawa H, Niimi N, Sango K, Nagai R. Glucoselysine, a unique advanced glycation end-product of the polyol pathway and its association with vascular complications in type 2 diabetes. J Biol Chem 2024; 300:107479. [PMID: 38879006 PMCID: PMC11283207 DOI: 10.1016/j.jbc.2024.107479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 07/13/2024] Open
Abstract
Glucoselysine (GL) is an unique advanced glycation end-product derived from fructose. The main source of fructose in vivo is the polyol pathway, and an increase in its activity leads to diabetic complications. Here, we aimed to demonstrate that GL can serve as an indicator of the polyol pathway activity. Additionally, we propose a novel approach for detecting GL in peripheral blood samples using liquid chromatography-tandem mass spectrometry and evaluate its clinical usefulness. We successfully circumvent interference from fructoselysine, which shares the same molecular weight as GL, by performing ultrafiltration and hydrolysis without reduction, successfully generating adequate peaks for quantification in serum. Furthermore, using immortalized aldose reductase KO mouse Schwann cells, we demonstrate that GL reflects the downstream activity of the polyol pathway and that GL produced intracellularly is released into the extracellular space. Clinical studies reveal that GL levels in patients with type 2 diabetes are significantly higher than those in healthy participants, while Nδ-(5-hydro-5-methyl-4-imidazolon-2-yl)ornithine (MG-H1) levels are significantly lower. Both GL and MG-H1 show higher values among patients with vascular complications; however, GL varies more markedly than MG-H1 as well as hemoglobin A1c, fasting plasma glucose, and estimated glomerular filtration rate. Furthermore, GL remains consistently stable under various existing drug treatments for type 2 diabetes, whereas MG-H1 is impacted. To the best of our knowledge, we provide important insights in predicting diabetic complications caused by enhanced polyol pathway activity via assessment of GL levels in peripheral blood samples from patients.
Collapse
Affiliation(s)
- Hiroko Yamaguchi
- Laboratory of Food and Regulation Biology, Graduate School of Bioscience, Tokai University, Kumamoto, Japan
| | - Takeshi Matsumura
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hikari Sugawa
- Laboratory of Food and Regulation Biology, Department of Food and Life Science, School of Agriculture, Tokai University, Kumamoto, Japan
| | - Naoko Niimi
- Diabetic Neuropathy Project, Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kazunori Sango
- Diabetic Neuropathy Project, Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Ryoji Nagai
- Laboratory of Food and Regulation Biology, Graduate School of Bioscience, Tokai University, Kumamoto, Japan; Laboratory of Food and Regulation Biology, Department of Food and Life Science, School of Agriculture, Tokai University, Kumamoto, Japan.
| |
Collapse
|
30
|
Korsmo HW, Ekperikpe US, Daehn IS. Emerging Roles of Xanthine Oxidoreductase in Chronic Kidney Disease. Antioxidants (Basel) 2024; 13:712. [PMID: 38929151 PMCID: PMC11200862 DOI: 10.3390/antiox13060712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Xanthine Oxidoreductase (XOR) is a ubiquitous, essential enzyme responsible for the terminal steps of purine catabolism, ultimately producing uric acid that is eliminated by the kidneys. XOR is also a physiological source of superoxide ion, hydrogen peroxide, and nitric oxide, which can function as second messengers in the activation of various physiological pathways, as well as contribute to the development and the progression of chronic conditions including kidney diseases, which are increasing in prevalence worldwide. XOR activity can promote oxidative distress, endothelial dysfunction, and inflammation through the biological effects of reactive oxygen species; nitric oxide and uric acid are the major products of XOR activity. However, the complex relationship of these reactions in disease settings has long been debated, and the environmental influences and genetics remain largely unknown. In this review, we give an overview of the biochemistry, biology, environmental, and current clinical impact of XOR in the kidney. Finally, we highlight recent genetic studies linking XOR and risk for kidney disease, igniting enthusiasm for future biomarker development and novel therapeutic approaches targeting XOR.
Collapse
Affiliation(s)
| | | | - Ilse S. Daehn
- Department of Medicine, Division of Nephrology, The Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, Box 1243, New York, NY 10029, USA
| |
Collapse
|
31
|
Alahmari H, Liu CC, Rubin E, Lin VY, Rodriguez P, Chang KC. Vitamin C alleviates hyperglycemic stress in retinal pigment epithelial cells. Mol Biol Rep 2024; 51:637. [PMID: 38727927 DOI: 10.1007/s11033-024-09595-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Retinal pigment epithelial cells (RPECs) are a type of retinal cells that structurally and physiologically support photoreceptors. However, hyperglycemia has been shown to play a critical role in the progression of diabetic retinopathy (DR), which is one of the leading causes of vision impairment. In the diabetic eye, the high glucose environment damages RPECs via the induction of oxidative stress, leading to the release of excess reactive oxygen species (ROS) and triggering apoptosis. In this study, we aim to investigate the antioxidant mechanism of Vitamin C in reducing hyperglycemia-induced stress and whether this mechanism can preserve the function of RPECs. METHODS AND RESULTS ARPE-19 cells were treated with high glucose in the presence or absence of Vitamin C. Cell viability was measured by MTT assay. Cleaved poly ADP-ribose polymerase (PARP) was used to identify apoptosis in the cells. ROS were detected by the DCFH-DA reaction. The accumulation of sorbitol in the aldose reductase (AR) polyol pathway was determined using the sorbitol detection assay. Primary mouse RPECs were isolated from adult mice and identified by Rpe65 expression. The mitochondrial damage was measured by mitochondrial membrane depolarization. Our results showed that high glucose conditions reduce cell viability in RPECs while Vitamin C can restore cell viability, compared to the vehicle treatment. We also demonstrated that Vitamin C reduces hyperglycemia-induced ROS production and prevents cell apoptosis in RPECs in an AR-independent pathway. CONCLUSIONS These results suggest that Vitamin C is not only a nutritional necessity but also an adjuvant that can be combined with AR inhibitors for alleviating hyperglycemic stress in RPECs.
Collapse
Affiliation(s)
- Hamid Alahmari
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Chia-Chun Liu
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Elizabeth Rubin
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Venice Y Lin
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
- North Allegheny Senior High School, Wexford, PA, 15090, USA
| | - Paul Rodriguez
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Kun-Che Chang
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA.
- Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
32
|
Rauf A, Naz S, Akram Z, Saleem M, Alomar TS, AlMasoud N, Ribaudo G. Bioactivity of 2-(3,4-Dihydroxyphenyl)-5,7-dihydroxy-2,3-dihydrochromen-4-one from pistacia chinensis on ENPP1 and aldose reductase. Nat Prod Res 2024:1-5. [PMID: 38712509 DOI: 10.1080/14786419.2024.2344744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/14/2024] [Indexed: 05/08/2024]
Abstract
Pistacia chinensis is used as a decorative tree and currently studied as a source of biofuels. Besides, its parts and extracts are endowed with several therapeutic uses which have been widely explored in traditional medicine and that are related to its rich composition in phytochemicals. Molecular docking and enzymatic inhibition tests were used to study the activity of eriodictyol, a flavonoid extracted from the barks of P. chinensis, against ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) and aldose reductase (ALR2). The compound was highlighted as a micromolar inhibitor in vitro (IC50 = 263.76 ± 1.32 µM and 4.21 ± 0.94 µM, respectively) and docking showed that eriodictyol efficiently targets the binding sites of the enzymes. In conclusion, this study unveils the potential of eriodictyol on enzymes that are involved in immunostimulation and in complications of diabetes mellitus.
Collapse
Affiliation(s)
- Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Pakistan
| | - Saima Naz
- Department of Biotechnology, Bacha Khan University Charsadda, Charsadda, Pakistan
| | - Zuneera Akram
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Baqai Medical University, Karachi, Pakistan
| | - Muhammad Saleem
- Department of Chemistry, Ghazi University, Dera Ghazi Khan, Pakistan
| | - Taghrid S Alomar
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Najla AlMasoud
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Giovanni Ribaudo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
33
|
Yasir M, Park J, Chun W. Discovery of Novel Aldose Reductase Inhibitors via the Integration of Ligand-Based and Structure-Based Virtual Screening with Experimental Validation. ACS OMEGA 2024; 9:20338-20349. [PMID: 38737046 PMCID: PMC11079907 DOI: 10.1021/acsomega.4c00820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/14/2024]
Abstract
Aldose reductase plays a central role in diabetes mellitus (DM) associated complications by converting glucose to sorbitol, resulting in a harmful increase of reactive oxygen species (ROS) in various tissues, such as the heart, vasculature, neurons, eyes, and kidneys. We employed a comprehensive approach, integrating both ligand- and structure-based virtual screening followed by experimental validation. Initially, candidate compounds were extracted from extensive drug and chemical libraries using the DeepChem's GraphConvMol algorithm, leveraging its capacity for robust molecular feature representation. Subsequent refinement employed molecular docking and molecular dynamics (MD) simulations, which are crucial for understanding compound-receptor interactions and dynamic behavior in a simulated physiological environment. Finally, the candidate compounds were subjected to experimental validation of their biological activity using an aldose reductase inhibitor screening kit. The comprehensive approach led to the identification of a promising compound, demonstrating significant potential as an aldose reductase inhibitor. This comprehensive approach not only yields a potential therapeutic intervention for DM-related complications but also establishes an integrated protocol for drug development, setting a new benchmark in the field.
Collapse
Affiliation(s)
- Muhammad Yasir
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Jinyoung Park
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| |
Collapse
|
34
|
Dmitrieva NI, Boehm M, Yancey PH, Enhörning S. Long-term health outcomes associated with hydration status. Nat Rev Nephrol 2024; 20:275-294. [PMID: 38409366 DOI: 10.1038/s41581-024-00817-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2024] [Indexed: 02/28/2024]
Abstract
Body water balance is determined by fundamental homeostatic mechanisms that maintain stable volume, osmolality and the composition of extracellular and intracellular fluids. Water balance is maintained by multiple mechanisms that continuously match water losses through urine, the skin, the gastrointestinal tract and respiration with water gains achieved through drinking, eating and metabolic water production. Hydration status is determined by the state of the water balance. Underhydration occurs when a decrease in body water availability, due to high losses or low gains, stimulates adaptive responses within the water balance network that are aimed at decreasing losses and increasing gains. This stimulation is also accompanied by cardiovascular adjustments. Epidemiological and experimental studies have linked markers of low fluid intake and underhydration - such as increased plasma concentration of vasopressin and sodium, as well as elevated urine osmolality - with an increased risk of new-onset chronic diseases, accelerated aging and premature mortality, suggesting that persistent activation of adaptive responses may be detrimental to long-term health outcomes. The causative nature of these associations is currently being tested in interventional trials. Understanding of the physiological responses to underhydration may help to identify possible mechanisms that underlie potential adverse, long-term effects of underhydration and inform future research to develop preventative and treatment approaches to the optimization of hydration status.
Collapse
Affiliation(s)
- Natalia I Dmitrieva
- Laboratory of Cardiovascular Regenerative Medicine, National Heart Lung and Blood Institute, NIH, Bethesda, Maryland, USA.
| | - Manfred Boehm
- Laboratory of Cardiovascular Regenerative Medicine, National Heart Lung and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Paul H Yancey
- Biology Department, Whitman College, Walla Walla, Washington, USA
| | - Sofia Enhörning
- Perinatal and Cardiovascular Epidemiology, Lund University Diabetes Centre, Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
- Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
35
|
Bekenova NB, Vochshenkova TA, Ablakimova N, Zhylkybekova A, Mussin NM, Albayev RK, Kaliyev AA, Tamadon A. A Bibliometric Analysis of Study of Associations of Certain Genotypes with the Cardiovascular Form of Diabetic Neuropathy. BIOMED RESEARCH INTERNATIONAL 2024; 2024:6761451. [PMID: 38659608 PMCID: PMC11042907 DOI: 10.1155/2024/6761451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/16/2024] [Accepted: 04/05/2024] [Indexed: 04/26/2024]
Abstract
This bibliometric analysis explores the landscape of research on the associations between specific genotypes and the cardiovascular form of diabetic neuropathy. Diabetes mellitus (DM) is a major contributor to premature mortality, primarily due to increased susceptibility to cardiovascular diseases. The global prevalence of DM is rising, with projections indicating further increases. Diabetic neuropathy, a complication of DM, includes the cardiovascular subtype, posing challenges in diagnosis and management. Understanding the genetic basis of cardiovascular diabetic neuropathy is crucial for targeted therapeutic interventions. The study utilizes bibliometric analysis to synthesize existing literature, identify trends, and guide future research. The Scopus database was searched, applying inclusion criteria for English articles related to genotypes and cardiovascular diabetic neuropathy. The analysis reveals a dynamic field with a notable impact, collaborative efforts, and multidimensional aspects. Publication trends over 1997-2023 demonstrate fluctuating research intensity. Top journals, authors, and affiliations are highlighted, emphasizing global contributions. Keyword analysis reveals thematic trends, and citation analysis identifies influential documents. Limitations include database biases, incomplete metadata, and search query specificity. The urgent need to explore genetic factors in cardiovascular diabetic neuropathy aligns with the increasing global diabetes burden. This analysis provides a comprehensive overview, contributing to the broader discourse on diabetic neuropathy research.
Collapse
Affiliation(s)
- Nazira B. Bekenova
- Gerontology Center, Medical Center of the President's Affairs Administration of the Republic of Kazakhstan, Astana, Kazakhstan
| | - Tamara A. Vochshenkova
- Gerontology Center, Medical Center of the President's Affairs Administration of the Republic of Kazakhstan, Astana, Kazakhstan
| | - Nurgul Ablakimova
- Department of Pharmacology, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Aliya Zhylkybekova
- Department of Evidence-Based Medicine and Scientific Management, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Nadiar M. Mussin
- General Surgery, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Rustam K. Albayev
- Gerontology Center, Medical Center of the President's Affairs Administration of the Republic of Kazakhstan, Astana, Kazakhstan
| | - Asset A. Kaliyev
- General Surgery, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Amin Tamadon
- Department for Natural Sciences, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
- PerciaVista R&D Co., Shiraz, Iran
| |
Collapse
|
36
|
Güleç Ö, Türkeş C, Arslan M, Demir Y, Dincer B, Ece A, İrfan Küfrevioğlu Ö, Beydemir Ş. Novel spiroindoline derivatives targeting aldose reductase against diabetic complications: Bioactivity, cytotoxicity, and molecular modeling studies. Bioorg Chem 2024; 145:107221. [PMID: 38387398 DOI: 10.1016/j.bioorg.2024.107221] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/01/2024] [Accepted: 02/16/2024] [Indexed: 02/24/2024]
Abstract
Despite significant developments in therapeutic strategies, Diabetes Mellitus remains an increasing concern, leading to various complications, e.g., cataracts, neuropathy, retinopathy, nephropathy, and several cardiovascular diseases. The polyol pathway, which involves Aldose reductase (AR) as a critical enzyme, has been focused on by many researchers as a target for intervention. On the other hand, spiroindoline-based compounds possess remarkable biological properties. This guided us to synthesize novel spiroindoline oxadiazolyl-based acetate derivatives and investigate their biological activities. The synthesized molecules' structures were confirmed herein, using IR, NMR (1H and 13C), and Mass spectroscopy. All compounds were potent inhibitors with KI constants spanning from 0.186 ± 0.020 μM to 0.662 ± 0.042 μM versus AR and appeared as better inhibitors than the clinically used drug, Epalrestat (EPR, KI: 0.841 ± 0.051 μM). Besides its remarkable inhibitory profile compared to EPR, compound 6k (KI: 0.186 ± 0.020 μM) was also determined to have an unusual pharmacokinetic profile. The results showed that 6k had less cytotoxic effect on normal mouse fibroblast (L929) cells (IC50 of 569.58 ± 0.80 μM) and reduced the viability of human breast adenocarcinoma (MCF-7) cells (IC50 of 110.87 ± 0.42 μM) more than the reference drug Doxorubicin (IC50s of 98.26 ± 0.45 μM and 158.49 ± 2.73 μM, respectively), thus exhibiting more potent anticancer activity. Moreover, molecular dynamic simulations for 200 ns were conducted to predict the docked complex's stability and reveal significant amino acid residues that 6k interacts with throughout the simulation.
Collapse
Affiliation(s)
- Özcan Güleç
- Department of Chemistry, Faculty of Arts and Sciences, Sakarya University, 54187 Sakarya, Turkey
| | - Cüneyt Türkeş
- Department of Biochemistry, Faculty of Pharmacy, Erzincan Binali Yıldırım University, 24002 Erzincan, Turkey.
| | - Mustafa Arslan
- Department of Chemistry, Faculty of Arts and Sciences, Sakarya University, 54187 Sakarya, Turkey.
| | - Yeliz Demir
- Department of Pharmacy Services, Nihat Delibalta Göle Vocational High School, Ardahan University, 75700 Ardahan, Turkey
| | - Busra Dincer
- Department of Pharmacology, Faculty of Pharmacy, Ondokuz Mayıs University, 55020 Samsun, Turkey
| | - Abdulilah Ece
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Biruni University, 34010 İstanbul, Turkey
| | | | - Şükrü Beydemir
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey; Bilecik Şeyh Edebali University, 11230 Bilecik, Turkey
| |
Collapse
|
37
|
Thakur MR, Nachane SS, Tupe RS. Alleviation of albumin glycation-induced diabetic cardiomyopathy by L-Arginine: Insights into Nrf-2 signaling. Int J Biol Macromol 2024; 264:130478. [PMID: 38428781 DOI: 10.1016/j.ijbiomac.2024.130478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024]
Abstract
In hyperglycemia, accelerated glycation and oxidative stress give rise to many diabetic complications, such as diabetic cardiomyopathy (DCM). Glycated human serum albumin (GHSA) has disturbed structural integrity and hampered functional capabilities. When GHSA accumulates around cardiac cells, Nrf-2 is dysregulated, aiding oxidative stress. L-Arginine (L-Arg) is prescribed to patients with diabetes and cardiovascular diseases. This research contributes to the mechanistic insights on antiglycation and antioxidant potential of L-Arg in alleviating DCM. HSA was glycated with methylglyoxal in the presence of L-Arg (20-640 mM). Structural and functional modifications of HSA were studied. L-Arg and HSA, GHSA interactions, and thermodynamics were determined by steady-state fluorescence. H9c2 cardiomyocytes were given treatments of GHSA-L-Arg along with the inhibitor of the receptor of AGEs. Cellular antioxidant levels, detoxification enzyme activities were measured. Gene, protein expressions, and immunofluorescence data examined the activation and nuclear translocation of Nrf-2 during glycation and oxidative stress. L-Arg protected HSA from glycation-induced structural and functional modifications. The binding affinity of L-Arg was more towards HSA (104 M-1). L-Arg, specifically at lower concentration (20 mM), upregulated Nrf-2 gene, protein expressions and facilitated its nuclear translocation by activating Nrf-2 signaling. The study concluded that L-Arg can be of therapeutic advantage in glycation-induced DCM and associated oxidative stress.
Collapse
Affiliation(s)
- Muskan R Thakur
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra State, India
| | - Sampada S Nachane
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra State, India
| | - Rashmi S Tupe
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra State, India.
| |
Collapse
|
38
|
Yang L, Xu L, Hao X, Song Z, Zhang X, Liu P, Wang S, He Z, Zou L. An aldose reductase inhibitor, WJ-39, ameliorates renal tubular injury in diabetic nephropathy by activating PINK1/Parkin signaling. Eur J Pharmacol 2024; 967:176376. [PMID: 38336014 DOI: 10.1016/j.ejphar.2024.176376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 01/18/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024]
Abstract
Renal tubular injury is a critical factor during the early stages of diabetic nephropathy (DN). Proximal tubular epithelial cells, which contain abundant mitochondria essential for intracellular homeostasis, are susceptible to disruptions in the intracellular environment, making them especially vulnerable to diabetic state disorders, which may be attributed to their elevated energy requirements and reliance on aerobic metabolism. It is widely thought that overactivation of the polyol pathway is implicated in DN pathogenesis, and inhibition of aldose reductase (AR), the rate-limiting enzyme in this pathway, represents a promising therapeutic avenue. WJ-39, a novel aldose reductase inhibitor, was investigated in this study for its protective effects on renal tubules in DN and the underlying mechanisms. Our findings revealed that WJ-39 significantly ameliorated the renal tubular morphology in high-fat diet (HFD)/streptozotocin (STZ)-induced DN rats, concurrently inhibiting fibrosis. Notably, WJ-39 safeguarded the structure and function of renal tubular mitochondria by enhancing mitochondrial dynamics. This involved the regulation of mitochondrial fission and fusion proteins and the promotion of PTEN-induced putative kinase 1 (PINK1)/Parkin-mediated mitophagy. Furthermore, WJ-39 demonstrated the inhibition of endogenous apoptosis by mitigating the production of mitochondrial reactive oxygen species (ROS). The protective effects of WJ-39 on mitochondria and apoptosis were countered in high glucose-treated HK-2 cells upon transfection with PINK1 siRNA. Overall, our findings suggest that WJ-39 protects the structural and functional integrity of renal tubules in DN, which may be attributed to its capacity to inhibit aldose reductase activity, activate the PINK1/Parkin signaling pathway, promote mitophagy, and alleviate apoptosis.
Collapse
Affiliation(s)
- Luxi Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Liangting Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Xin Hao
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Zhixiao Song
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Xian Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Peng Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Shaojie Wang
- Department of Pharmacochemistry, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang Liaoning, 110016, China.
| | - Zhonggui He
- Department of Pharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China.
| | - Libo Zou
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China.
| |
Collapse
|
39
|
Mohammed FS, Babu D, Irfan Z, Fayed MA. A review on the traditional uses, nutritive importance, pharmacognostic features, phytochemicals, and pharmacology of Momordica cymbalaria Hook F. PeerJ 2024; 12:e16928. [PMID: 38436002 PMCID: PMC10906271 DOI: 10.7717/peerj.16928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 01/22/2024] [Indexed: 03/05/2024] Open
Abstract
Momordica cymbalaria Hook F. (MC), belonging to the family Cucurbitaceae, is a plant with several biological activities. This detailed, comprehensive review gathers and presents all the information related to the geographical distribution, morphology, therapeutic uses, nutritional values, pharmacognostic characters, phytochemicals, and pharmacological activities of MC. The available literature showed that MC fruits are utilized as a stimulant, tonic, laxative, stomachic, and to combat inflammatory disorders. The fruits are used to treat spleen and liver diseases and are applied in folk medicine to induce abortion and treat diabetes mellitus. The phytochemical screening studies report that MC fruits contain tannins, alkaloids, phenols, proteins, amino acids, vitamin C, carbohydrates, β-carotenes, palmitic acid, oleic acid, stearic acid, α-eleostearic acid, and γ-linolenic acid. The fruits also contain calcium, sodium, iron, potassium, copper, manganese, zinc, and phosphorus. Notably, momordicosides are cucurbitacin triterpenoids reported in the fruits of MC. Diverse pharmacological activities of MC, such as analgesic, anti-inflammatory, antioxidant, hepatoprotective, nephroprotective, antidiabetic, cardioprotective, antidepressant, anticonvulsant, anticancer, antiangiogenic, antifertility, antiulcer, antimicrobial, antidiarrheal and anthelmintic, have been reported by many investigators. M. cymbalaria methanolic extract is safe up to 2,000 mg/kg. Furthermore, no symptoms of toxicity were found. These pharmacological activities are mechanistically interpreted and described in this review. Additionally, the microscopic, powder and physiochemical characteristics of MC tubers are also highlighted. In summary, possesses remarkable medicinal values, which warrant further detailed studies to exploit its potential benefits therapeutically.
Collapse
Affiliation(s)
- Firdous Sayeed Mohammed
- Department of Pharmacology, Calcutta Institute of Pharmaceutical Technology and Allied Health Sciences, Uluberia, Howrah, West Bengal, India
| | - Dinesh Babu
- Faculty of Pharmacy and Pharmaceutical Sciences, Katz Group Centre for Pharmacy and Health Research, University of Alberta, Alberta, Canada
| | - Zainab Irfan
- Department of Pharmaceutical Technology, Brainware University, Kolkata, West Bengal, India
| | - Marwa A.A. Fayed
- Pharmacognosy Department, Faculty of Pharmacy, University of Sadat City, Sadat City, Egypt
| |
Collapse
|
40
|
Syamprasad NP, Jain S, Rajdev B, Panda SR, Kumar GJ, Shaik KM, Shantanu P, Challa VS, Jorvekar SB, Borkar RM, Vaidya JR, Tripathi DM, Naidu V. AKR1B1 drives hyperglycemia-induced metabolic reprogramming in MASLD-associated hepatocellular carcinoma. JHEP Rep 2024; 6:100974. [PMID: 38283757 PMCID: PMC10820337 DOI: 10.1016/j.jhepr.2023.100974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 10/21/2023] [Accepted: 10/25/2023] [Indexed: 01/30/2024] Open
Abstract
Background & Aims The mechanism behind the progressive pathological alteration in metabolic dysfunction-associated steatotic liver disease/steatohepatitis (MASLD/MASH)-associated hepatocellular carcinoma (HCC) is poorly understood. In the present study, we investigated the role of the polyol pathway enzyme AKR1B1 in metabolic switching associated with MASLD/MASH and in the progression of HCC. Methods AKR1B1 expression was estimated in the tissue and plasma of patients with MASLD/MASH, HCC, and HCC with diabetes mellitus. The role of AKR1B1 in metabolic switching in vitro was assessed through media conditioning, lentiviral transfection, and pharmacological probes. A proteomic and metabolomic approach was applied for the in-depth investigation of metabolic pathways. Preclinically, mice were subjected to a high-fructose diet and diethylnitrosamine to investigate the role of AKR1B1 in the hyperglycemia-mediated metabolic switching characteristic of MASLD-HCC. Results A significant increase in the expression of AKR1B1 was observed in tissue and plasma samples from patients with MASLD/MASH, HCC, and HCC with diabetes mellitus compared to normal samples. Mechanistically, in vitro assays revealed that AKR1B1 modulates the Warburg effect, mitochondrial dynamics, the tricarboxylic acid cycle, and lipogenesis to promote hyperglycemia-mediated MASLD and cancer progression. A pathological increase in the expression of AKR1B1 was observed in experimental MASLD-HCC, and expression was positively correlated with high blood glucose levels. High-fructose diet + diethylnitrosamine-treated animals also exhibited statistically significant elevation of metabolic markers and carcinogenesis markers. AKR1B1 inhibition with epalrestat or NARI-29 inhibited cellular metabolism in in vitro and in vivo models. Conclusions Pathological AKR1B1 modulates hepatic metabolism to promote MASLD-associated hepatocarcinogenesis. Aldose reductase inhibition modulates the glycolytic pathway to prevent precancerous hepatocyte formation. Impact and implications This research work highlights AKR1B1 as a druggable target in metabolic dysfunction-associated steatotic liver disease (MASLD) and hepatocellular carcinoma (HCC), which could provide the basis for the development of new chemotherapeutic agents. Moreover, our results indicate the potential of plasma AKR1B1 levels as a prognostic marker and diagnostic test for MASLD and associated HCC. Additionally, a major observation in this study was that AKR1B1 is associated with the promotion of the Warburg effect in HCC.
Collapse
Affiliation(s)
- NP Syamprasad
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila village, Changsari, Assam, 781101, India
| | - Siddhi Jain
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila village, Changsari, Assam, 781101, India
| | - Bishal Rajdev
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila village, Changsari, Assam, 781101, India
| | - Samir Ranjan Panda
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila village, Changsari, Assam, 781101, India
| | - Gangasani Jagadeesh Kumar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila village, Changsari, Assam, 781101, India
| | - Khaja Moinuddin Shaik
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila village, Changsari, Assam, 781101, India
| | - P.A. Shantanu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila village, Changsari, Assam, 781101, India
| | - Veerabhadra Swamy Challa
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila village, Changsari, Assam, 781101, India
| | - Sachin B. Jorvekar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research Guwahati, Sila village, Changsari, Assam, 781101, India
| | - Roshan M. Borkar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research Guwahati, Sila village, Changsari, Assam, 781101, India
| | - Jayathirtha Rao Vaidya
- Fluoro Agro Chemicals Department and AcSIR-Ghaziabad, CSIR-Indian Institute of Chemical Technology, Uppal Road Tarnaka, Hyderabad, Telangana, 500007, India
| | - Dinesh Mani Tripathi
- Liver Physiology & Vascular Biology Lab, Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, ILBS, D-1, Vasant Kunj, New Delhi, Delhi 110070, India
| | - V.G.M. Naidu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila village, Changsari, Assam, 781101, India
| |
Collapse
|
41
|
Awote OK, Kanmodi RI, Ebube SC, Abdulganniyyu ZF. Nutritional Profile, GC-MS Analysis and In-silico Anti-diabetic Phytocompounds Candidature of Jatropha gossypifolia Leaf Extracts. Curr Drug Discov Technol 2024; 21:32-45. [PMID: 37817655 DOI: 10.2174/0115701638267143230925172207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/17/2023] [Accepted: 08/31/2023] [Indexed: 10/12/2023]
Abstract
BACKGROUND Diabetes mellitus (DM) is a metabolic disorder known to impair many physiological functions via reactive oxygen species (ROS). Aldose reductase, sorbitol dehydrogenase, dipeptidyl peptidase IV, α-amylase and α-glucosidase are pharmacotherapeutic protein targets in type-2 diabetes mellitus (T2DM). Inhibitors of these enzymes constitute a new class of drugs used in the management and treatment of T2DM. Some reports have claimed that medicinal plant extracts that serves as food (and as an antioxidant source) can reduce these alterations by eliminating ROS caused by DM. Ethnobotanical survey claims Jatropha gossypifolia commonly called "fignut" and "Lapa-lapa" in the Yoruba land of South-western Nigeria, to be used for the treatment and management of diabetes, in addition to its nutritive value. OBJECTIVE The nutritional composition and in-silico antidiabetic potential of the bioactive constituents of J. gossypifolia leaf extracts were investigated. METHODS Proximate, minerals and gas chromatography-mass spectroscopy (GC-MS) analysis were carried out using standard procedures. Phytocompounds present in J. gossypifolia methanol (JGM) and ethyl acetate (JGE) leaf extracts were tested as potential antagonists of selected protein targets via in-silico techniques. Drug-likeness, pharmacokinetic properties and toxicity of the promising docked ligands were also predicted. RESULTS The proximate and mineral analysis revealed good nutritional composition and mineral content. Additionally, cyclo-pentadecane and dibutyl phthalate from methanol extract, and benzene- 1,2,4,5-tetramethyl, benzene-1,2,3,5-tetramethyl, and benzene-1,3-dimethyl-5-(1-methylethyl) from ethyl acetate extract were present in J. gossypifolia leaf which exhibited a better binding affinity than the clinically prescribed standard, metformin. CONCLUSION Benzene-1,2,4,5-tetramethyl from JGE extracts exhibited the most promising antidiabetic potential in-silico, suggesting its candidature as diabetes-target-protein inhibitor which may be developed for the treatment of type-2 diabetes mellitus.
Collapse
Affiliation(s)
- Olasunkanmi Kayode Awote
- Department of Biochemistry, Faculty of Science, Lagos State University, Ojo, Lagos State, Nigeria
| | - Rahmon Ilesanmi Kanmodi
- Department of Biochemistry, Faculty of Science, Lagos State University, Ojo, Lagos State, Nigeria
| | - Success Chidera Ebube
- Department of Biochemistry, Faculty of Science, Lagos State University, Ojo, Lagos State, Nigeria
| | | |
Collapse
|
42
|
Liao Y, Mao H, Gao X, Lin H, Li W, Chen Y, Li H. Drug screening identifies aldose reductase as a novel target for treating cisplatin-induced hearing loss. Free Radic Biol Med 2024; 210:430-447. [PMID: 38056576 DOI: 10.1016/j.freeradbiomed.2023.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/08/2023] [Accepted: 11/24/2023] [Indexed: 12/08/2023]
Abstract
Cisplatin is a frequently used chemotherapeutic medicine for cancer treatment. Permanent hearing loss is one of the most serious side effects of cisplatin, but there are few FDA-approved medicines to prevent it. We applied high-through screening and target fishing and identified aldose reductase, a key enzyme of the polyol pathway, as a novel target for treating cisplatin ototoxicity. Cisplatin treatment significantly increased the expression level and enzyme activity of aldose reductase in the cochlear sensory epithelium. Genetic knockdown or pharmacological inhibition of aldose reductase showed a significant protective effect on cochlear hair cells. Cisplatin-induced overactivation of aldose reductase led to the decrease of NADPH/NADP+ and GSH/GSSG ratios, as well as the increase of oxidative stress, and contributed to hair cell death. Results of target prediction, molecular docking, and enzyme activity detection further identified that Tiliroside was an effective inhibitor of aldose reductase. Tiliroside was proven to inhibit the enzymatic activity of aldose reductase via competitively interfering with the substrate-binding region. Both Tiliroside and another clinically approved aldose reductase inhibitor, Epalrestat, inhibited cisplatin-induced oxidative stress and subsequent cell death and thus protected hearing function. These findings discovered the role of aldose reductase in the pathogenesis of cisplatin-induced deafness and identified aldose reductase as a new target for the prevention and treatment of hearing loss.
Collapse
Affiliation(s)
- Yaqi Liao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, PR China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, PR China; ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, PR China
| | - Huanyu Mao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, PR China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, PR China; ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, PR China
| | - Xian Gao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, PR China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, PR China; ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, PR China
| | - Hailiang Lin
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, PR China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, PR China; ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, PR China
| | - Wenyan Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, PR China; Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, PR China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, PR China; The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, PR China; ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, PR China.
| | - Yan Chen
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, PR China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, PR China; ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, PR China.
| | - Huawei Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, PR China; Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, PR China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, PR China; The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, PR China; ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, PR China.
| |
Collapse
|
43
|
Siddique R, Mehmood MH, Shehzad MA. Current antioxidant medicinal regime and treatments used to alleviate oxidative stress in infertility issues. FUNDAMENTAL PRINCIPLES OF OXIDATIVE STRESS IN METABOLISM AND REPRODUCTION 2024:287-315. [DOI: 10.1016/b978-0-443-18807-7.00018-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
44
|
Jiang R, Zhang H, Liu Q, Yang X, He L, Yuan L, Cheng D. De Novo Design of Near-Infrared Fluorescent Agents Activated by Peroxynitrite and Glutathione-Responsive Imaging for Diabetic Liver Disease. Adv Healthc Mater 2024; 13:e2302466. [PMID: 37840532 DOI: 10.1002/adhm.202302466] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/11/2023] [Indexed: 10/17/2023]
Abstract
Diabetes and its complications, such as diabetes liver disease, is a major problem puzzling people's health. The detection of redox states in its pathological process can effectively help us gain a deeper understanding of the disease. The pair of oxidation-reduction substances peroxynitrite (ONOO- ) and glutathione (GSH) is considered to be closely related to their occurrence and development. Thus, direct visualization of ONOO- and GSH in diabetes liver disease is critical to evaluate the disease at the molecular level. Herein, two activatable agents NTCF-ONOO- and NTCF-GSH are prepared for selectively detecting ONOO- and GSH through protection and deprotection strategies based on hydroxyl and amino groups of near-infrared fluorophore. Fluorescence imaging of exogenous and endogenous ONOO- and GSH changes in living cells and in vivo is observed. The ONOO- and GSH level in the diabetes liver disease cellular model are visualized and the possible redox imbalance mechanism related to the oxidized (NAD+ ) and reduced (NADH) nicotinamide adenine dinucleotides is explored in this process. Moreover, these probes can sensitively recognize ONOO- and GSH in the process of oxidative stress resulting from streptozotocin and streptozotocin/acetaminophen-induced complex diabetic liver disease in vivo. In addition, they can be applied for monitoring the clinical serum sample related with diabetic patients.
Collapse
Affiliation(s)
- Renfeng Jiang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Hongshuai Zhang
- Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver Disease, Clinical Research Institute, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Qian Liu
- Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver Disease, Clinical Research Institute, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Xuefeng Yang
- Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver Disease, Clinical Research Institute, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Longwei He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Lin Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Dan Cheng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
- Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver Disease, Clinical Research Institute, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| |
Collapse
|
45
|
Chen S, Zou Y, Song C, Cao K, Cai K, Wu Y, Zhang Z, Geng D, Sun W, Ouyang N, Zhang N, Li Z, Sun G, Zhang Y, Sun Y, Zhang Y. The role of glycolytic metabolic pathways in cardiovascular disease and potential therapeutic approaches. Basic Res Cardiol 2023; 118:48. [PMID: 37938421 PMCID: PMC10632287 DOI: 10.1007/s00395-023-01018-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/09/2023]
Abstract
Cardiovascular disease (CVD) is a major threat to human health, accounting for 46% of non-communicable disease deaths. Glycolysis is a conserved and rigorous biological process that breaks down glucose into pyruvate, and its primary function is to provide the body with the energy and intermediate products needed for life activities. The non-glycolytic actions of enzymes associated with the glycolytic pathway have long been found to be associated with the development of CVD, typically exemplified by metabolic remodeling in heart failure, which is a condition in which the heart exhibits a rapid adaptive response to hypoxic and hypoxic conditions, occurring early in the course of heart failure. It is mainly characterized by a decrease in oxidative phosphorylation and a rise in the glycolytic pathway, and the rise in glycolysis is considered a hallmark of metabolic remodeling. In addition to this, the glycolytic metabolic pathway is the main source of energy for cardiomyocytes during ischemia-reperfusion. Not only that, the auxiliary pathways of glycolysis, such as the polyol pathway, hexosamine pathway, and pentose phosphate pathway, are also closely related to CVD. Therefore, targeting glycolysis is very attractive for therapeutic intervention in CVD. However, the relationship between glycolytic pathway and CVD is very complex, and some preclinical studies have confirmed that targeting glycolysis does have a certain degree of efficacy, but its specific role in the development of CVD has yet to be explored. This article aims to summarize the current knowledge regarding the glycolytic pathway and its key enzymes (including hexokinase (HK), phosphoglucose isomerase (PGI), phosphofructokinase-1 (PFK1), aldolase (Aldolase), phosphoglycerate metatase (PGAM), enolase (ENO) pyruvate kinase (PKM) lactate dehydrogenase (LDH)) for their role in cardiovascular diseases (e.g., heart failure, myocardial infarction, atherosclerosis) and possible emerging therapeutic targets.
Collapse
Affiliation(s)
- Shuxian Chen
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cai
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Zhaobo Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Wei Sun
- Department of Thyroid Surgery, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Nanxiang Ouyang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Naijin Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
- Key Laboratory of Reproductive and Genetic Medicine, China Medical University, National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Zhao Li
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Guozhe Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| |
Collapse
|
46
|
Liu Y, Wang D, Liu YP. Metabolite profiles of diabetes mellitus and response to intervention in anti-hyperglycemic drugs. Front Endocrinol (Lausanne) 2023; 14:1237934. [PMID: 38027178 PMCID: PMC10644798 DOI: 10.3389/fendo.2023.1237934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) has become a major health problem, threatening the quality of life of nearly 500 million patients worldwide. As a typical multifactorial metabolic disease, T2DM involves the changes and interactions of various metabolic pathways such as carbohydrates, amino acid, and lipids. It has been suggested that metabolites are not only the endpoints of upstream biochemical processes, but also play a critical role as regulators of disease progression. For example, excess free fatty acids can lead to reduced glucose utilization in skeletal muscle and induce insulin resistance; metabolism disorder of branched-chain amino acids contributes to the accumulation of toxic metabolic intermediates, and promotes the dysfunction of β-cell mitochondria, stress signal transduction, and apoptosis. In this paper, we discuss the role of metabolites in the pathogenesis of T2DM and their potential as biomarkers. Finally, we list the effects of anti-hyperglycemic drugs on serum/plasma metabolic profiles.
Collapse
Affiliation(s)
| | | | - Yi-Ping Liu
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou, China
| |
Collapse
|
47
|
Di Benedetto C, Borini Etichetti C, Cocordano N, Cantoia A, Arel Zalazar E, Bicciato S, Menacho-Márquez M, Rosano GL, Girardini J. The p53 tumor suppressor regulates AKR1B1 expression, a metastasis-promoting gene in breast cancer. Front Mol Biosci 2023; 10:1145279. [PMID: 37780210 PMCID: PMC10538543 DOI: 10.3389/fmolb.2023.1145279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 08/28/2023] [Indexed: 10/03/2023] Open
Abstract
Alteration of metabolism in cancer cells is a central aspect of the mechanisms that sustain aggressive traits. Aldo-keto reductase 1 B1 (AKR1B1) catalyzes the reduction of several aldehydes to alcohols consuming NADPH. Nevertheless, the ability of AKR1B1 to reduce different substrates renders difficult to comprehensively ascertain its biological role. Recent evidence has implicated AKR1B1 in cancer; however, the mechanisms underlying its pro-oncogenic function remain largely unknown. In this work, we report that AKR1B1 expression is controlled by the p53 tumor suppressor. We found that breast cancer patients bearing wild-type TP53 have reduced AKR1B1 expression. In cancer cell lines, p53 reduced AKR1B1 mRNA and protein levels and repressed promoter activity in luciferase assays. Furthermore, chromatin immunoprecipitation assays indicated that p53 is recruited to the AKR1B1 promoter. We also observed that AKR1B1 overexpression promoted metastasis in the 4T1 orthotopic model of triple-negative breast cancer. Proteomic analysis of 4T1 cells overexpressing AKR1B1 showed that AKR1B1 exerts a marked effect on proteins related to metabolism, with a particular impact on mitochondrial function. This work provides novel insights on the link between the p53 pathway and metabolism in cancer cells and contributes to characterizing the alterations associated to the pathologic role of AKR1B1.
Collapse
Affiliation(s)
- Carolina Di Benedetto
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, United States
| | - Carla Borini Etichetti
- Instituto de Fisiología Experimental de Rosario (IFISE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Nabila Cocordano
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Alejo Cantoia
- Unidad de Espectrometría de Masa, Instituto de Biología Molecular y Celular de Rosario (IBR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Evelyn Arel Zalazar
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Mauricio Menacho-Márquez
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Germán Leandro Rosano
- Unidad de Espectrometría de Masa, Instituto de Biología Molecular y Celular de Rosario (IBR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Javier Girardini
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| |
Collapse
|
48
|
Singh Y, Kumar N, Kulkarni S, Singh S, Thareja S. Pharmacophore derived 3D-QSAR, molecular docking, and simulation studies of quinoxaline derivatives as ALR2 inhibitors. J Biomol Struct Dyn 2023; 42:10452-10488. [PMID: 37698364 DOI: 10.1080/07391102.2023.2256885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/04/2023] [Indexed: 09/13/2023]
Abstract
Aldose Reductase 2 (ALR2), a key enzyme of the polyol pathway, plays a crucial role in the pathogenesis of diabetic complications. Quinoxaline scaffold-based compounds have been identified as potential ALR2 inhibitors for the management of diabetic complications. In the present work, molecular dynamic simulation studies in conjugation with pharmacophore mapping and atom-based 3D-QSAR were performed on a dataset of 99 molecules in comparison with Epalrestat (reference) to mark the desirable structural features of quinoxaline analogs to generate a probable template for designing novel and effective ALR2 inhibitors. The most potent compound 81 was subjected to MD simulation studies and found to be stable, with better interactions with the binding pocket as compared to Epalrestat. The MM-GBSA and MM-PBSA calculations showed that compound 81 possessed binding free energies of -35.96 and -4.92 kcal/mol, respectively. Atom-based 3D-QSAR yielded various pharmacophoric features with excellent statistical measures, such as correlation coefficient (R2 value), F-value (Fischer ratio), Q2 value (cross-validated correlation coefficient), and Pearson's R-value for training and test sets. Furthermore, the pharmacophore mapping provided a five-point hypothesis (AADRR) and docking analysis revealed the active ligand-binding orientations on the active site's amino acid residues TYR 48, HIE 110, TRP 111, and TRP 219. The results of this study will help in designing potent inhibitors of ALR2 for the management of diabetic complications.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yogesh Singh
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Niraj Kumar
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Swanand Kulkarni
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Satwinder Singh
- Department of Computer Science and Technology, Central University of Punjab, Bathinda, India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab, Bathinda, India
| |
Collapse
|
49
|
Chen M, Chen Y, Zhu W, Yan X, Xiao J, Zhang P, Liu P, Li P. Advances in the pharmacological study of Chinese herbal medicine to alleviate diabetic nephropathy by improving mitochondrial oxidative stress. Biomed Pharmacother 2023; 165:115088. [PMID: 37413900 DOI: 10.1016/j.biopha.2023.115088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/08/2023] Open
Abstract
Diabetic nephropathy (DN) is one of the serious complications of diabetes mellitus, primarily arising from type 2 diabetes (T2DM), and can progress to chronic kidney disease (CKD) and end stage renal disease (ESRD). The pathogenesis of DN involves various factors such as hemodynamic changes, oxidative stress, inflammatory response, and lipid metabolism disorders. Increasing attention is being given to DN caused by oxidative stress in the mitochondrial pathway, prompting researchers to explore drugs that can regulate these target pathways. Chinese herbal medicine, known for its accessibility, rich historical usage, and remarkable efficacy, has shown promise in ameliorating renal injury caused by DN by modulating oxidative stress in the mitochondrial pathway. This review aims to provide a reference for the prevention and treatment of DN. Firstly, we outline the mechanisms by which mitochondrial dysfunction impairs DN, focusing on outlining the damage to mitochondria by oxidative stress. Subsequently, we describe the process by which formulas, herbs and monomeric compounds protect the kidney by ameliorating oxidative stress in the mitochondrial pathway. Finally, the rich variety of Chinese herbal medicine, combined with modern extraction techniques, has great potential, and as we gradually understand the pathogenesis of DN and research techniques are constantly updated, there will be more and more promising therapeutic targets and herbal drug candidates. This paper aims to provide a reference for the prevention and treatment of DN.
Collapse
Affiliation(s)
- Ming Chen
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Yao Chen
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Wenhui Zhu
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Xiaoming Yan
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Jing Xiao
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Peiqing Zhang
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China.
| | - Peng Liu
- Shunyi Hospital, Beijing Hospital of Traditional Chinese Medicine, Beijing, China.
| | - Ping Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
50
|
Cancemi G, Cicero N, Allegra A, Gangemi S. Effect of Diet and Oxidative Stress in the Pathogenesis of Lymphoproliferative Disorders. Antioxidants (Basel) 2023; 12:1674. [PMID: 37759977 PMCID: PMC10525385 DOI: 10.3390/antiox12091674] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/19/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Lymphomas are a heterogeneous group of pathologies that result from clonal proliferation of lymphocytes. They are classified into Hodgkin lymphoma and non-Hodgkin lymphoma; the latter develops as a result of B, T, or NK cells undergoing malignant transformation. It is believed that diet can modulate cellular redox state and that oxidative stress is implicated in lymphomagenesis by acting on several biological mechanisms; in fact, oxidative stress can generate a state of chronic inflammation through the activation of various transcription factors, thereby increasing the production of proinflammatory cytokines and causing overstimulation of B lymphocytes in the production of antibodies and possible alterations in cellular DNA. The purpose of our work is to investigate the results of in vitro and in vivo studies on the possible interaction between lymphomas, oxidative stress, and diet. A variety of dietary regimens and substances introduced with the diet that may have antioxidant and antiproliferative effects were assessed. The possibility of using nutraceuticals as novel anticancer agents is discussed; although the use of natural substances in lymphoma therapy is an interesting field of study, further studies are needed to define the efficacy of different nutraceuticals before introducing them into clinical practice.
Collapse
Affiliation(s)
- Gabriella Cancemi
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (G.C.); (A.A.)
| | - Nicola Cicero
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (G.C.); (A.A.)
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| |
Collapse
|